Skip to main content

Full text of "USPTO Patents Application 10551504"

See other formats


(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) 



(19) World Intellectual Property Organization 

International Bureau 

(43) International Publication Date 
11 October 2001 (11.10.2001) 




PCT 



(10) International Publication Number 

WO 01/74388 Al 



(51) International Patent Classification 7 : A61K 39/395, 
A61P 35/00 // C07K 16/24, 16/28, (A61K 39/395, 31:00) 

(21) International Application Number: PCT/US01/10382 

(22) International Filing Date: 2 April 2001 (02.04.2001) 

(25) Filing Language: English 

(26) Publication Language: English 

(30) Priority Data: 

60/193,467 31 March 2000 (31.03.2000) US 

(71) Applicant: IDEC PHARMACEUTICALS CORPORA- 
TION [US/US]; 11011 Torreyana Road, San Diego, CA 
92121 (US). 

(72) Inventor: HANNA, Nabil; 14770 Avenida Insurgentes, 
ft= Rancho Santa Fe, CA 92067 (US). 

^= (74) Agents: TESKIN, Robin, L. et al.; Pillsbury Winthrop 
^5 LLP, 1100 New York Avenue, N.W., Washington, DC 
5= 20005 (US). 



(81) Designated States (national): AE, AG, AL, AM, AT, AU, 
AZ, BA, BB, BG, BR, BY, BZ, CA, CH, CN, CO, CR, CU, 
CZ, DE, DK, DM, DZ, EE, ES, FI, GB, GD, GE, GH, GM, 
HR, HU, ID, IL, IN, IS, JP, KE, KG, KP, KR, KZ, LC, LK, 
LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, MX, 
MZ, NO, NZ, PL, PT, RO, RU, SD, SE, SG, SI, SK, SL, 
TJ, TM, TR, TT, TZ, UA, UG, UZ, VN, YU, ZA, ZW. 

(84) Designated States (regional): ARIPO patent (GH, GM, 
KE, LS, MW, MZ, SD, SL, SZ, TZ, UG, ZW), Eurasian 
patent (AM, AZ, BY, KG, KZ, MD, RU, TJ, TM), European 
patent (AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, 
IT, LU, MC, NL, PT, SE, TR), OAPI patent (BF, BJ, CF, 
CG, CI, CM, GA, GN, GW, ML, MR, NE, SN, TD, TG). 

Published: 

— with international search report 

— before the expiration of the time limit for amending the 
claims and to be republished in the event of receipt of 
amendments 

For two-letter codes and other abbreviations, refer to the "Guid- 
ance Notes on Codes and Abbreviations" appearing at the begin- 
ning of each regular issue of the PCT Gazette. 



qq (54) Title: COMBINED USE OF ANTT-CYTOKINE ANTIBODIES OR ANTAGONISTS AND ANTI-CD20 FOR THE TREAT- 
00 MENT OF B CELL LYMPHOMA 

|J (57) Abstract: The present invention discloses combined therapies for treating hematologic malignancies, including B cell lym- 
***-; phomas and leukemias or solid non-hematologic tumors, comprising administration of anti-cytokine antibodies or antagonists to 
^ inhibit the activity of cytokines which play a role in perpetuating the activation of B cells. The administration of such antibodies 
and antagonists, particularly anti-ILlO antibodies and antagonists, is particularly useful for avoiding or decreasing the resistance of 
Q hematologic malignant cells or solid tumor cells to chemotherapeutic agents and anti-CD20 or anti-CD22 antibodies. The invention 
^ also provides combination therapies for solid tumors having B cell involvement comprising the administration of an anti-cytokine 
^ antibody and a B cell depleting antibody such as RITUSAN®. 



WO 01/74388 



PCT/US01/10382 



COMBINED USE OF ANTI-CYTOKINE ANTIBODIES 
OR ANTAGONISTS AND ANTI-CD20 FOR THE TREATMENT 
OF B CELL LYMPHOMA 

5 FIELD OF THE INVENTION 

The present invention concerns methods for treating hematologic malignancies 
including B cell lymphomas and leukemias with anti-cytokine agents such as 
antibodies and antagonists, where the targeted cytokines play a potentiating role in the 
disease process by stimulating hematologic malignant cells including B lymphoma 

10 and leukemia cells. Treatment with anti-cytokine agents in combination with other 
known therapies such as chemotherapy and administration of therapeutic antibodies 
has been found to provide a synergistic effect. 

The invention also embraces the treatment of solid non-hematologic (non- 
lymphoid) tumors, e.g., colorectal or liver cancer, which tumors are characterized by 

15 B cell involvement, by the administration of a cytokine antibody or cytokine 

antagonist, in combination with treatment with an antibody to a B cell target, e.g. 
CD20. 

BACKGROUND OF THE INVENTION 

20 The immune system of vertebrates (for example, primates, which include humans, 
apes, monkeys, etc.) consists of a number of organs and cell types which have evolved 
to: accurately and specifically recognize foreign microorganisms ("antigen") which 
invade the vertebrate-host; specifically bind to such foreign microorganisms; and, 
eliminate/destroy such foreign microorganisms. Lymphocytes, as well as other types 

25 of cells, are critical to the immune system and to the elimination and destruction of 
foreign microorganisms. Lymphocytes are produced in the thymus, spleen and bone 
marrow (adult) and represent about 30% of the total white blood cells present in the 
circulatory system of humans (adult). There are two major sub-populations of 
lymphocytes: T cells and B cells. T cells are responsible for cell mediated immunity, 

30 while B cells are responsible for antibody production (humoral immunity). However, 
T cells and B cells can be considered interdependent — in a typical immune response, 
T cells are activated when the T cell receptor binds to fragments of an antigen that are 
bound to major histocompatability complex ("MHC") glycoproteins on the surface of 
an antigen presenting cell; such activation causes release of biological mediators 



WO 01/74388 



PCTAJS01/10382 



("interleukins" or "cytokines") which, in essence, stimulate B cells to differentiate and 
produce antibody ("immunoglobulins") against the antigen. 

Each B cell within the host expresses a different antibody on its surface- thus 
one B cell will express antibody specific for one antigen, while another B cell will 
5 express antibody specific for a different antigen. Accordingly, B cells are quite 
diverse, and this diversity is critical to the immune system. In humans, each B cell 
can produce an enormous number of antibody molecules (i.e., about 10 7 to 10 8 ). Such 
antibody production most typically ceases (or substantially decreases) when the 
foreign antigen has been neutralized. Occasionally, however, proliferation of a 

10 particular B cell will continue unabated; such proliferation can result in a cancer 
referred to as "B cell lymphoma." 

Non-Hodgkin's lymphoma is one type of lymphoma that is characterized by the 
malignant growth of B lymphocytes. According to the American Cancer Society, an 
estimated 54,000 new cases will be diagnosed, 65% of which will be classified as 

15 intermediate- or high-grade lymphoma. Patients diagnosed with intermediate-grade 
lymphoma have an average survival rate of two to five years, and patients diagnosed 
with high-grade lymphoma survive an average of six months to two years after 
diagnosis. 

Conventional therapies have included chemotherapy and radiation, possibly 
20 accompanied by either autologous or allogeneic bone marrow or stem cell 

transplantation if a suitable donor is available, and if the bone marrow contains too 
many tumor cells upon harvesting. While patients often respond to conventional 
therapies, they usually relapse within several months. 

A relatively new approach to treating non-Hodgkin's lymphoma has been to 
25 treat patients with a monoclonal antibody directed to a protein on the surface of 
cancerous B cells. The antibody may be conjugated to a toxin or radiolabel thereby 
affecting cell death after binding. Alternatively, an antibody may be engineered with 
human constant regions such that human antibody effector mechanisms are generated 
upon antibody binding which result in apoptosis or death of the cell. 



WO 01/74388 



PCT/US01/10382 



Rituximab® (DEC Pharmaceuticals Corporation) is one of a new generation 
of monoclonal antibodies developed for the treatment of B cell lymphomas, and in 
particular, non-Hodgkin's lymphoma. Rituximab® is a genetically engineered anti- 
CD20 monoclonal antibody with murine light-and heavy-chain variable regions and 
5 human gamma I heavy-chain and kappa light-chain constant regions. Rituximab® is 
more effective than its murine parent in fixing complement and mediating ADCC, and 
it mediates CDC in the presence of human complement. The antibody inhibits cell 
growth in the B-cell lines FL-18, Ramos, and Raji, sensitizes chemoresistant human 
lymphoma cell lines to diphtheria toxin, ricin, CDDP, doxorubicin, and etoposide, and 
10 induces apoptosis in the DHL-4 human B-cell lymphoma line in a dose-dependent 
manner. 

However, many patients are refractory to or relapse following Rituximab® 
therapy, as well as chemotherapy. Therefor, there still remains a need for lymphoma 
treatments which may be combined with Rituximab® therapy or chemotherapy in 
15 order to increase the chance of remission and decrease the rate of relapse in 
lymphoma patients. 

Many groups have suggested using cytokines for the treatment of various types 
of cancers. For instance, Wang et al. suggested that cytokines are "directly cytotoxic 
to tumor cells" and showed that interleukin-1 alpha (ILla) potentiated the anti-tumor 

20 effect of anti-tumor drugs against several human tumor cells in vitro (Int. J. Cancer 
(Nov. 27, 1996) 68(5): 583-587). Bonvida et al. disclose that cytokines have the 
potential to "enhance the efficacy of chemotherapeutic agents" and show that 
recombinant tumor necrosis factor and the chemotherapeutic agent cisplatin show a 
synergistic effect against ovarian cancer cells (Gynecol. Oncol. (Sept. 1990) 38(3): 

25 333-339). U.S. Patent No. 5,716,612 teaches that IL-4 may be used to potentiate the 
effect of chemotherapeutic agents in the treatment of cancer. 

However, some groups have also recognized that cytokines may play a 
detrimental role in the development of some cancers. For instance, interleukin-6 (IL6) 
has been known for the ability in some instances to inhibit apoptosis of leukemic 

30 cells. (See Yonish-Rouach et al. Wild type p53 induces apoptosis of myeloid 



WO 01/74388 PCT/US01/10382 

leukemic cells and is inhibited by interleukin-6. Nature 352: 345-347 (1991)). 
Recently it was shown that JL6 may play a role in the resistance of some leukemic 
cells to anti-cancer chemotherapeutic agents, and that, in vitro, anti-IL6 antibody 
increases the sensitivity of cisplatin-resistance K562 cells to cisp latin-induced 
5 apoptosis. (See Dedoussis et al. Endogenous interleukin 6 conveys resistance to cis- 
diamminedichloroplatinum-mediated apoptosis of the K562 human leukemic cell 
line). 

A potentiating effect on B cells has also been postulated for IL10, the 
production of which has been reported to be upregulated in some cell lines derived 

10 from B cell lymphomas (See Cortes et al. Interleukin- 10 in non-Hodgkin's 

lymphoma. Leuk Lymphoma 26(3-4): 251-259 (July, 1997). However, when the 
serum of NHL patients was tested for correlation between IL10 levels and prognosis, 
more significance was placed on the levels of viral IL10, which is produced from a 
homologous open reading frame BCFR1, located in the genome of the Epstein Barr 

15 Virus (EBV). In fact, another group reported at about the same time that IL10 was an 
autocrine growth factor for EBV-infected lymphoma cells. (See Beatty et al. 
Involvement of EL10 in the autonomous growth of EBV-transformed B cell lines. J. 
Immunol. 158(9): 4045-51 (May 1, 1997)). Alternatively, others have hypothesized 
that EL6 and IL10 production by macrophages plays a key role in the occurrence of 

20 lymphocytic diseases. {See U.S. Patent No. 5,639,600). 

It has also been reported that IL10 may work in combination with IL6, IL2 and 
TNF-alpha to increase proliferation of non-Hodgkin's lymphoma cells. (See 
Voorzanger et al. Interleukin-(IL)10 and IL6 are produced in vivo by non-Hodgkin's 
lymphoma cells and act as cooperative growth factors. Cancer Res. 56(23): 5499-505 

25 (Dec. 1, 1996). Also statistically significantly higher levels of IL2, IL6, IL8, IL10, 

soluble IL2 receptor, soluble transferrin receptor and neopterin were observed in NHL 
patients as compared to a control group, although no single parameter was found to be 
of prognostic significance. {See Stasi et al. Clinical implications of cytokine and 
soluble receptor measurements in patients with newly diagnosed non-Hodgkin's 

30 lymphoma. Eur. J. Haemotol. 54(1): 9-17 (Jan., 1995). 



-4- 



WO 01/74388 PCT/US01/10382 

However, there have been j ust as many reports in the literature which have 
suggested that cytokines such as IL10 show no correlation to disease progression, and 
that such cytokines may actually be helpful in combating lymphoma rather than 
contributing to the disease. For instance, when Bonnefoix et al. tested the potential of 
5 ten cytokines (JL2, TL3, IL4, TL6, IL10, IL13, G-CSF, GM-CSF, interferon alpha and 
interferon gamma) to modulate the spontaneous proliferative response of B-non- 
Hodgkin's lymphoma cells of various histological subtypes, this group found that 
each cytokine could be either inhibitory or stimulatory depending on the sample, and 
that there was no relationship with different histological subtypes, m fact, U.S. Patent 

1 0 No. 5,770, 1 90, herein incorporated by reference, suggests administration of IL1 0 in 
conjunction with chemotherapeutic agents as a treatment for acute leukemia. 

It would be a benefit to lymphoma patients if therapeutic regimens 
incorporating anti-cytokine antibodies could be devised whereby such antibodies 
could be used to increase the sensitivity of B lymphoma cells to other types of 

1 5 therapeutic drugs . It would be particularly helpful if anti-cytokine antibodies could be 
administered for the purpose of avoiding or overcoming the resistance of B lymphoma 
cells in lymphoma patients to chemotherapeutic agents, and for the purpose of 
potentiating the apoptotic activity of .therapeutic antibodies. Such combined treatment 
regimens would add to the therapies available to lymphoma patients and potentially 

20 decrease the rate of relapse in these patients. 

OBJECTS OF THE INVENTION 

It is an object of the invention to provide a method of avoiding, decreasing or 
overcoming the resistance of hematologic malignant cells or solid non-hematologic 
25 tumor cells to at least one chemotherapeutic agent, comprising administering an anti- 
cytokine antibody or cytokine antagonist to a patient diagnosed with a hematologic 
malignancy prior, concurrent or after administration of at least one chemotherapeutic 
agent. 

It is a more specific object of the invention to provide a method of avoiding, 
30 decreasing or overcoming the resistance of hematologic malignant cells to apoptosis 



-5- 



WO 01/74388 



PCT/US01/10382 



induced by a therapeutic agent, comprising administering an anti-cytokine antibody or 
cytokine antagonist to a patient diagnosed with a hematologic malignancy. 

It is another object of the invention to provide a method of treating a patient 
with a hematologic malignancy who has relapsed following chemotherapy, 
5 comprising administering an anti-cytokine antibody or cytokine antagonist to said 
patient. 

It is another object of the invention to provide a method of treating a patient 
having a hematologic malignancy who is refractory to chemotherapy, comprising 
administering an anti-cytokine antibody or cytokine antagonist to said patient. 

10 It is yet another object of the invention to provide a method of treating a 

patient with a hematologic malignancy who has relapsed following therapy with a 
therapeutic antibody, comprising administering an anti-cytokine antibody or cytokine 
antagonist to said patient. 

It is still another object of the invention to provide a method of treating a 

15 patient with a hematologic malignancy who is refractory to therapy with a therapeutic 
antibody, comprising administering an anti-cytokine antibody or cytokine antagonist 
to said patient. 

It is another object of the invention to provide a method of treating a B cell 
lymphoma patient comprising administering to said patient a therapeutically effective 
20 amount of an anti-CD20 antibody simultaneously with or consecutively with in either 
order an anti-cytokine antibody. 

It is another object of the invention to provide a method of treating a solid 
non-hematologic (non-lymphoid) tumor wherein B cells elicit a pro-tumor response 
by the adniinistration of an anti-cytokine antibody, e.g. an anti-ILlO antibody and at 
25 least one B cell depleting antibody, e.g. an anti-CD20 antibody. 

It is a more specific object of the invention to provide a method of treating 
solid, non-lymphoid tumor involving the digestive system, especially colorectal 
cancer or liver cancer by the administration of an anti-cytokine antibody, preferably an 
anti-ILlO antibody and a B cell depleting antibody, particularly a depleting anti-CD20 
30 antibody. 



-6- 



WO 01/74388 



PCT/US01/10382 



SUMMARY OF THE INVENTION 

In a first aspect, the present invention relates to the administration of anti- 
cytokine antibodies and cytokine antagonists, particularly antibodies to IL10, in 
5 combination with chemotherapy drugs and/or therapeutic antibodies to increase the 
response rate and response duration in patients with hematological malignancies such 
as B cell lymphomas and leukemias or solid non-hematologic tumors, such as breast 
cancer, ovarian cancer, testicular cancer and others. Thus, the present invention 
relates to methods of treating hematologic malignancies such as B cell lymphomas 

1 0 and leukemias by administering to a patient having a hematologic malignancy such as 
B cell lymphoma or a leukemia, antibodies directed to B cell receptors and antibodies 
or antagonists which interfere with the action of certain cytokines. In particular, the 
present invention relates to administration of antibodies to B cell markers which 
initiate apoptosis of B lymphoma cells, such as anti-CD20, anti-CD22, anti-CD40, 

15 anti-CD23, anti-CD19, anti-CD37 and others identified infra, and antibodies to or 
antagonists of cytokines which may interfere with apoptosis, e.g., anti-lLlO. 
Combined therapeutic regimens including other treatments which would also benefit 
from anti-cytokine therapy, i.e., chemotherapy, are also encompassed. The methods 
will find use in particular for treating patients having hematological malignancies 

20 such as B lymphomas or leukemias characterized by cells that have become resistant 
to chemotherapeutic agents and therapeutic antibodies. 

In a second aspect, the present invention provides novel methods of treating 
solid non-hematologic (non-lymphoid) tumors having B cell involvement (but not of 
B cell origin), particularly cancers wherein B cells elicit a pro-tumor response by the 

25 adrninistration of an antibody to a cytokine, e.g., IL10, in conjunction with B cell 
specific antibody therapy, particularly a B cell depleting antibody, and preferably 
CD20 antibody therapy, optionally in combination with radiotherapy or 
chemotherapy. Examples of such solid tumors include colorectal cancer, liver cancer, 
breast cancer, lung cancer, prostate cancer, stomach cancer, head and neck cancer, 

30 ovarian cancer, testicular cancer, esophageal cancer and others. Suitable 



-7- 



WO 01/74388 PCT/US01/10382 

chemotherapies are discussed infra. These cancers may comprise precancers, Stage I 
and H cancers, and advanced cancers, e.g. past Stage II and including solid tumors that 
have metastasized. 

5 DETAILED DESCRIPTION OF THE INVENTION 

In a first aspect, the present invention includes methods of avoiding, 
decreasing or overcoming the resistance of hematologic malignant cells including, 
e.g., B lymphoma and leukemia cells to at least one chemotherapeutic agent, 
comprising administering an anti-cytokine antibody or cytokine antagonist to a patient 

1 0 diagnosed with B cell lymphoma. 

Often, such resistance by a hematologic malignancy patient's B cells is 
mediated by stimulation of the tumorigenic B cells by one or more cytokines such that 
the cells fail to respond to apoptotic signals. In such cases, the methods of the present 
invention may be described as methods of avoiding, decreasing or overcoming the 

1 5 resistance of such tumorigenic B cells to apoptosis, with chemotherapeutic agents 
being examples of agents which may induce apoptosis. Also encompassed are 
therapeutic antibodies directed to targets on the surface of B cells, such as anti-CD 19, 
anti-CD20, anti-CD22, anti-CD40, and anti-CD28 and other B cell targets identified 
infra. 

20 Because resistance of B cells is often only apparent after a patient has relapsed 

following, or is refractory to, a first treatment with a therapeutic agent, the methods of 
the present invention will often encompass treating patients with hematologic 
malignancies such as B cell lymphoma or leukemia who have relapsed following, or 
are refractory to, chemotherapy or therapy with a therapeutic antibody. However, the 

25 anti-cytokine antibodies and antagonists of the present invention may also be used in 
conjunction with other therapies or prior to other therapies in patients newly 
diagnosed with lymphoma to decrease the chance of relapse, and increase the length 
and duration of the response to therapy. 

The methods of the present invention are appropriate to treat a wide variety of 

30 hematologic malignancies, especially B cell lymphomas and leukemias, including but 



-8- 



WO 01/74388 PCT/US01/10382 

not limited to low grade/follicular non-Hodgkin's lymphoma (NHL), small 
lymphocytic (SL) NHL, intermediate grade/ follicular NHL, intermediate grade 
diffuse NHL, high grade immunoblastic NHL, high grade lymphoblastic NHL, high 
grade small non-cleaved cell NHL, bulky disease NHL and Waldenstrom's 
5 MacroglobuUnemia, chronic leukocytic leukemia, acute myelogenous leukemia, acute 
lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous 
leukemia, lymphoblastic leukemia, lymphocytic leukemia, monocytic leukemia, 
myelogenous leukemia, and promyelocytic leukemia. It should be clear to those of 
skill in the art that these lymphomas will often have different names due to changing 

10 systems of classification, and that patients having lymphomas and leukemias 
classified under different names may also benefit from the combined therapeutic 
regimens of the present invention. 

For instance, a recent classification system proposed by European and 
American pathologists is called the Revised European American Lymphoma (REAL) 

15 Classification. This classification system recognizes Mantle cell lymphoma and 
Marginal cell lymphoma among other peripheral B-cell neoplasms, and separates 
some classifications into grades based on cytology, i.e., small cell, mixed small and 
large, large cell. It will be understood that all such classified lymphomas may benefit 
from the combined therapies of the present invention. 

20 The U.S. National Cancer Institute (NCI) has in turn divided some of the 

REAL classes into more clinically useful "indolent" or "aggressive" lymphoma 
designations. Indolent lymphomas include follicular cell lymphomas, separated into 
cytology "grades," diffuse small lymphocytic lymphoma/ chronic lymphocytic 
leukemia (CLL), lymphoplasmacytoidV Waldenstrom's Macroglobulinemia, Marginal 

25 zone lymphoma and Hairy cell leukemia. Aggressive lymphomas include diffuse 
mixed and large cell lymphoma, Burkitt's lymphoma/ diffuse small non-cleaved cell 
lymphoma, Lymphoblastic lymphoma, Mantle cell lymphoma and AIDS-related 
lymphoma. All that is required is that the extent or duration of response to therapy be 
extended as a result of administration of said anti-cytokine antibody or antagonist. 

30 But the methods are most preferably used to treat patients having non-Hodgkin's 



-9- 



WO 01/74388 PCT/US01/10382 



lymphoma (NHL), where the present inventors have surprisingly found that 
administration of anti-cytokine antibodies and antagonists has a synergistic effect. 
Since the effect of cytokines and the identity of detrimental cytokines may vary 
among different patients and different types of lymphomas, and the effect of various 
5 cytokines on the resistance of B lymphoma cells may vary with different 

chemotherapeutic and immunotherapeutic agents, it is suggested that the levels of the 
respective cytokines in individual patients be tested before the patients are 
administered the anti-cytokine therapy. 

In a second aspect, the invention provides a method of treating solid, non- 
10 hematologic tumors wherein B cells elicit a protein response (promote tumor growth 
and/or metastasis) comprising the admimstration of a anti-cytokine antibody, e.g. an 
anti-ILlO antibody, and an antibody to a B cell target, preferably an anti-CD20 
antibody having B cell depleting activity. However, the invention includes the usage 
of antibodies to other B cell targets identified infra. Also, this aspect further includes 
15 the additional use of chemotherapy and/or radiotherapy. 

A variety of chemotherapeutic agents have been applied to the treatment of 
different types of cancers, and the methods of the present invention will avoid, 
decrease or overcome the resistance of malignant, e.g. lymphoma, cells to at least one, 
but possibly several, of these chemotherapeutic agents. In particular, chemotherapies 
20 which may benefit by supplemental anti-cytokine therapy include but are not limited 
to CHOP, ICE, Mitozantrone, Cytarabine, DVP, ATRA, Idarubicin, hoelzer 
chemotherapy regime, La La chemotherapy regime, ABVD, CEOP, 2-CdA, FLAG & 
IDA with or without subsequent G-CSF treatment), VAD, M & P, C-Weekly, ABCM, 
MOPP, DHAP, methotrexate, doxorubicin, daunorubicin, tamoxifen, toremifene, and 
25 cisplatin. Other chemotherapeutic agents are identified infra in the section relating to 
preferred embodiments. 

There are likely to be a variety of cytokines which play a detrimental, 
stimulatory role in hematologic or non-hematologic malignancies including leukemic 
and lymphoma diseases, either alone or in cooperation with other cytokines. Thus, 
30 depending on the patient and the disease, more than one anti-cytokine antibody or 



-10- 



WO 01/74388 



PCT/US01/10382 



antagonist may benefit a particular patient as a supplemental therapy. Those 
cytokines include but are not limited to IL2, IL6, IL10 and TNF-alpha. Other 
appropriate cytokines are identified infra in the preferred embodiments. For non- 
Hodgkin's lymphoma, the preferred anti-cytokine treatment will comprise anti-ILlO 
5 therapy. 

There are several anti-ILlO antibodies which are known in the art and may be 
used for the purposes of the present invention. U.S. Patent No. 5,871,725 describes a 
rat anti-human antibody designated 19F1. Another anti-ILlO antibody, alpha-ILlO, is 
described in U.S. Patent No. 5,837,293. Anti-ILlO antibodies are also described in 

10 Tim R. Mosmann, et al., "Isolation of Monoclonal Antibodies Specific For IL-4, IL-5, 
IL-6, and a New Th2-Specific Cytokine (TL-1 0), Cytokine Synthesis Inhibitory Factor, 
By Using A Solid Phase Radiohmmmoadsorbent Assay," The Journal of Immunology, 
145(9):2938-2945, Nov. 1, 1990. Antagonists may take the form of proteins which 
compete for receptor binding, e.g., which lack the ability to activate the receptor while 

15 blocking IL-10 binding, or IL-10 binding molecules, such as antibodies. The term 
antibody should be understood as encompassing antibody fragments as well as whole 
antibodies, i.e., Fab, Fab 2 and Fv fragments. Antibodies may be isolated by 
immunizing another animal with human IL-10, but then may be humanized using 
method known in the art to decrease their immunogenicity once they are administered 

20 to a human patient. 

The appropriate dosage of anti-cytokine antibody will depend on the cytokine 
targeted, the results of preliminary serum profiles in individual patients, the type of 
lymphoma being treated and the stage of disease. For anti-ILlO antibodies in the 
treatment of newly diagnosed low-grade non-Hodgkin's lymphoma, the preferred 

25 dosage may range from .001 mg to 100 mg/kg, preferably from about 0.1 to 100 

mg/kg, and most typically about 0.4 to 20 mg/kg body weight, depending on whether 
the antibody is administered concurrently with or prior to another therapeutic agent. 
Preferably, the anti-cytokine antibody is administered concurrent or prior to a 
chemotherapeutic agent or other therapy, typically from about one hour prior, to about 



- 11- 



WO 01/74388 



PCIYUS01/10382 



one month prior, preferably within one to seven days prior to administration of 
chemotherapeutic or other agent. 

Also included in the present invention are kits for accomphshing the disclosed 
methods. A kit according to the present invention comprises at least one anti-cytokine 
5 antibody or antagonist which may be readily admixed or resuspended with a 
pharmaceutically acceptable carrier and conveniently injected into a lymphoma 
patient. In cases where the serum of a lymphoma patient is preferably tested for 
cytokine profiles prior to administration of said anti-cytokine antibody or antagonist, 
the kit may also or alternatively comprise reagents and materials for testing the 

10 relative amounts of various cytokines in the patient's serum. 

Also encompassed in the present invention are combined therapeutic methods 
of treating hematologic malignancies such as B cell lymphoma and leukemias 
comprising administering to a patient with a hematologic malignancy a therapeutically 
effective amount of a therapeutic antibody simultaneously with or consecutively with 

15 in either order an anti-cytokine antibody. Therapeutic antibodies are defined as those 
which bind to receptors on the surface of hematologic malignant cells, e.g., 
tumorigenic B cells, and mediate their destruction or depletion when they bind, i.e., 
anti-CD20, anti-CD19, anti-CD22, anti-CD21, anti-CD23, anti-CD37, and other B 
cell targets identified infra. While the anti-cytokine agents of the present invention 

20 will have some beneficial effect alone in that they block cytokine-mediated 

proliferation of tumorigenic B cells, the combined administration of the therapeutic 
antibodies with the anti-cytokine agents will have a synergistic effect in that the 
duration and/or extent of response will be better than the additive effect of both types 
of therapies applied independently. 

25 While not wishing to be held to the following theory, the present inventors 

believe that the synergistic effects seen by co-administering the anti-cytokine agents 
of the present invention are related to the inhibition of the targeted cytokine which 
may usually have the effect to inhibit apoptosis. Accordingly, when the anti-cytokine 
agents of the present invention are combined with an agent which acts by inducing 

30 apoptosis, e.g., anti-CD20, anti-CD22, anti-CD19, anti-CD21, anti-CD23, or anti- 



-12- 



WO 01/74388 



PCT/US01/10382 



CD40 antibodies, the combined administration shows a synergetic effect well beyond 
the additive effect of either agent alone. 

Never-the-less, this does not preclude the use of the anti-cytokine antibodies 
and antagonists of the present invention in combined therapies with other antibodies 
5 or therapeutic agents whose efficacy is not facilitated via apoptosis. For instance, 
radiolabeled antibodies facilitate the destruction of tumor cells by binding to the B 
cell surface and delivering a lethal dose of radiation. Such antibodies, as well as 
antibodies conjugated to toxins, may also be used in conjunction with the anti- 
cytokine agents of the present invention. Preferred radiolabeled antibodies are those 

1 0 labeled with yttrium-[90] C°Y). A particularly preferred radiolabeled antibody is 
Zevelin (IDEC Pharmaceuticals Corporation), which is an anti-CD20 antibody 
conjugated to 90 Y. 

The combined therapeutic methods of the present invention may further 
comprise administration of at least one chemotherapeutic agent or regimen, where 

1 5 such chemotherapy includes, by way of example, CHOP, ICE, Mitozantrone, 
Cytarabine, DVP, ATRA, Idarubicin, hoelzer chemotherapy regime, La La 
chemotherapy regime, ABVD, CEOP, 2-CdA, FLAG & IDA with or without 
subsequent G-CSF treatment), VAD, M & P, C-Weekly, ABCM, MOPP, DHAP, 
doxorubicin, cisplatin, daunorubicin, tamoxifen, toremifene, and methotrexate as well 

20 as the additional chemotherapeutic agents identified infra. A preferred 

chemotherapeutic regimen for the treatment of non-Hodgkin's lymphoma patients is 
CHOP. The anti-cytokine antibody or antagonist is preferably administered prior to 
the B cell target antibody, e.g., anti-CD20, CD22, CD 19 or CD40, and/or 
chemotherapy, such that proliferation of B lymphoma cells as a result of the targeted 

25 cytokine is quelled prior to administration of the B cell therapeutic. As described 
above, the target cytokine may be IL2, IL6, IL10 or TNF-alpha among others, 
depending on the patient's cytokine profile prior to treatment, but preferably the 
targeted cytokine is IL10. 

As mentioned above, therapeutic antibodies of the present invention may be 

30 any antibody which targets a molecule expressed on the surface of B cells, particularly 



- 13- 



WO 01/74388 PCT/US01/10382 

one having B cell depleting activity. A listing of suitable B cell targets is identified 
infra. 

Depending on the patient and extent of disease, the anti-B cell target binding 
antibody, e.g., Rituximab® may be administered at a dosage ranging from .01 to about 
5 100 mg/kg, more preferably from about .1 to 50 mg/kg, and most preferably from 
about .4 to 20 mg/kg of body weight. Effective dosages may be lower in combined 
therapeutic regimens which include anti-cytokine agents, because the proliferative 
potential of B lymphoma cells will be reduced. Again, effective doses will depend on 
the chosen anti-cytokine therapy, and the relative levels of potentiating cytokine in the 

10 patient's serum. 

The combined therapies of the present invention are also suitable for treating a 
wide range of lymphomas, including but not limited to low grade/follicular non- 
Hodgkin's lymphoma (NHL), small lymphocytic (SL) NHL, intermediate grade/ 
follicular NHL, intermediate grade diffuse NHL, high grade immunoblastic NHL, 

1 5 high grade lymphoblastic NHL, high grade small non-cleaved cell NHL, bulky disease 
NHL and Waldenstrom's Macroglobulinemia, chronic leukocytic leukemia, acute 
myelogenous leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, 
chronic myelogenous leukemia, lymphoblastic leukemia, lymphocytic leukemia, 
monocytic leukemia, myelogenous leukemia, and promyelocytic leukemia Preferred 

20 targeted diseases are non-Hodgkin's lymphoma (NHL), and particularly low-grade, 
follicular NHL. Again, it may be helpful for the serum of the lymphoma patient to be 
tested for cytokine profiles prior to administration of the anti-cytokine antibody or 
antagonist. 

As already discussed, the combination therapies provided herein, particularly 
25 the combined usage of an anti-cytokine antibody, e.g. anti-ILlO and an anti-B cell 
target antibody, e.g. anti-CD20, are also useful for treating solid, non-hematologic 
(non-lymphoid) cancers, including by way of example, colorectal cancer, liver cancer, 
and other digestive cancers, breast cancer, esophageal cancer, head and neck cancer, 
lung cancer, ovarian cancer, prostate cancer and testicular cancer. These cancers my 
30 be in early, intermediate or advanced stages, e.g. metastasis. 



-14- 



WO 01/74388 



PCT/US01/10382 



The present invention also encompasses kits for administering the therapeutic 
antibody and the anti-cytokine antibody or antagonist according to the disclosed 
methods. Kits may comprise more than one type of therapeutic antibody and more 
than one anti-cytokine agent. Kits may also comprise reagents and materials for 
5 testing cytokine profile prior to administration of the therapeutic antibody and anti- 
cytokine antibody or antagonist. 

As noted, the invention further embraces the treatment of solid, non-lymphoid 
tumors by the administration of an anti-cytokine antibody, e.g., an anti-ILlO antibody, 
and a B cell specific antibody, preferably an antibody having substantial B cell 
1 0 depleting activity such as RITUXAN® . It ahs been reported that some solid tumors 
apparently have B cell involvement. That is to say that the B cells are somehow 
involved in promoting or mamtaining the tumorigenic state and may impede the 
body's immune defense system against such tumor. With respect thereto, WO 020864 
Al, incorporated by reference herein, which identifies Biocrystal Inc. as the Applicant 
1 5 describes the treatment of solid, non-lymphoid tumor using antibodies that target B 
cells, including Rituxan®. It was reported therein that this treatment resulted in 
pronounced anti-tumor responses, even in patients with advanced colorectal cancer, 
lung cancer and liver cancer. 

By contrast, the present invention provides an improved combination therapy, 
20 wherein solid, non-lymphoid tumors are treated by use of an anti-cytokine antibody, 
such as anti-ILlO and a B cell depleting antibody, such as an anti-CD20 antibody. 

This combination regimen should afford an enhanced method of treating solid 
tumors, particularly those wherein B cells are involved, but are not themselves the 
cancerous cells. In this regimen, the cytokine antagonist, e.g., anti-cytokine antibody 
25 and the B cell depleting antibody, e.g., Rituxan® will be administered separately or 
together and in either order. 

Additionally, this regimen may include the use of radiotherapy, e.g., external 
beam irradiation, total body irradiation, ra<noimmunotherapy or chemotherapy. 
Suitable chemotherapies are identified infra. The radioimmunotherapy may comprise 



-15- 



WO 01/74388 PCT/US01/10382 

treatment with a radiolabeled antibody that binds a target expressed by the solid 
tumor. 

Typically, the anti-cytokine antibody will be administered prior to the B cell 
depleting antibody. It is anticipated that this combination therapy will be suitable for 
5 treating any solid tumor having B cell involvement. Suitable examples of solid 

tumors have been identified previously. One noteworthy example is colorectal cancer. 

In this embodiment, the B cell depleting antibody and cytokine will be 
administered such that it s delivered to the solid tumor site. Preferably, the antibodies 
will be injected proximate or directly at the tumor site, e.g., by intravenous injection at 
10 a vein proximate to the tumor. 

This combination regimen cell results in remission or shrinkage of the solid 
tumor, e.g., a lung or colorectal tumor. 

DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS 

15 In order to further describe the preferred embodiments and full scope of the 

invention, the following definitions are provided. 
I. Definitions 

"Cytokine antagonist" is a compound that inhibits or blocks the expression 
and/or activity of a cytokine, e.g. an interleukin or interferon or another cytokine. 

20 A "B cell surface marker" or "B cell target" or "B cell antigen" herein is an 

antigen expressed on the surface of a B cell which can be targeted with an antagonist 
which binds thereto. Exemplary B cell surface markers include the CD10, CD19, 
CD20, CD21, CD22, CD23, CD24, CD37, CD53, CD72, CD73, CD74, CDw75, 
CDw76, CD77, CDw78, CD79a, CD79b, CD80, CD81, CD82, CD83, CDw84, CD85 

25 and CD86 leukocyte surface markers: The B cell surface marker of particular interest 
is preferentially expressed on B cells compared to other non-B cell tissues of a 
mammal and may be expressed on both precursor B cells and mature B cells. In one 
embodiment, the marker is one, like CD20 or CD 19, which is found on B cells 
throughout differentiation of the lineage from the stem cell stage up to a point just 

30 prior to terminal differentiation into plasma cells. The preferred B cell surface 



- 16- 



WO 01/74388 



PCTAJS01/10382 



markers herein are CD19 and CD20. 

The "CD20" antigen is a - 35 kDa, non-glycosylated phosphoprotein found on 
the surface of greater than 90% of B cells from peripheral blood or lymphoid organs. 
CD20 is expressed during early pre-B cell development and remains until plasma cell 
5 differentiation. CD20 is present on both normal B cells as well as malignant B cells. 
Other names for CD20 in the literature include "B-lymphocyte-restricted antigen" and 
"Bp35". The CD20 antigen is described in Clark et al. PNAS (USA) 82:1766 (1985), 
for example. The "CD19" antigen refers to a -90kDa antigen identified, for example, 
by the HD237-CD19 or 134 antibody (Kiesel et al. Leukemia Research 11, 12: 1 1 19 
10 (1987)). Like CD20, CD19 is found on cells throughout differentiation of the lineage 
from the stem cell stage up to a point just prior to terminal differentiation into plasma 
cells. Binding of an antagonist to CD 19 may cause internalization of the CD 19 
antigen. 

A "hematologic malignancy" includes any malignancy associated with cells in 

15 the bloodstream. Examples thereof include B and T cell lymphomas, leukemias 
including but not limited to low grade/follicular non-Hodgkin's lymphoma (NHL), 
small lymphocytic (SL) NHL, intermediate grade/ follicular NHL, intermediate grade 
diffuse NHL, high grade immunoblastic NHL, high grade lymphoblastic NHL, high 
grade small non-cleaved cell NHL, bulky disease NHL and Waldenstrom's 

20 Macroglobulinemia, chronic leukocytic leukemia, acute myelogenous leukemia, acute 
lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous 
leukemia, lymphoblastic leukemia, lymphocytic leukemia, monocytic leukemia, 
myelogenous leukemia, and promyelocytic leukemia. It should be clear to those of 
skill in the art that these lymphomas will often have different names due to changing 

25 systems of classification (as previously discussed), and that patients having 

lymphomas and leukemias classified under different names may also benefit from the 
combined therapeutic regimens of the present invention. 

A solid, non-hematologic (non-lymphoid) tumor refers to a non-hematologic 
malignancy having B cell involvement, i.e., where B cells are involved in a 

30 "protumor" response. Such solid tumors are characterized by palpable tumors, 



-17- 



WO 01/74388 PCTYUS01/10382 

typically at least 0.5 mm in diameter, more typically at least 1 .0 mm in diameter. 
Examples thereof include colorectal cancer, liver cancer, breast cancer, lung cancer, 
head and neck cancer, stomach cancer, testicular cancer, prostate cancer, ovarian 
cancer, uterine cancer and others. These cancers may be in the early stages 
5 (precancer), intermediate (Stages I and II) or advanced, including solid tumors that 
have metastasized. These solid tumors will preferably be cancers wherein B cells 
elicit a protumor response, i.e. the presence of B cells is involved in tumor 
development, maintenance or metastasis. 

A B cell "antagonist" is a molecule which, upon binding to a B cell surface 

10 marker, destroys or depletes B cells in a mammal and/or interferes with one or more B 
cell functions, e.g. by reducing or preventing a humoral response elicited by the B 
cell. The antagonist preferably is able to deplete B cells (i.e. reduce circulating B cell 
levels) in a mammal treated therewith. Such depletion may be achieved via various 
mechanisms such antibody-dependent cell mediated cytotoxicity (ADCC) and/or 

1 5 complement dependent cytotoxicity (CDC), inhibition of B cell proliferation and/or 
induction of B cell death (e.g. via apoptosis). Antagonists included within the scope of 
the present invention include antibodies, synthetic or native sequence peptides and 
small molecule antagonists which bind to the B cell marker, optionally conjugated 
with or fused to a cytotoxic agent. The preferred antagonist comprises an antibody, 

20 more preferably a B cell depleting antibody. 

"Antibody-dependent cell-mediated cytotoxicity" and "ADCC" refer to a 
cell-mediated reaction in which nonspecific cytotoxic cells that express Fc receptors 
(FcRs) (e.g. Natural Killer (NK) cells, neutrophils, and macrophages) recognize 
bound antibody on a target cell and subsequently cause lysis of the target cell. The 

25 primary cells for mediating ADCC, NK cells, express FcyRIH only, whereas 

monocytes express FcyRI, FcyRII and FcyRHI. FcR expression on hematopoietic 
cells in summarized is Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. 
Immunol 9:457-92 (1991). To assess ADCC activity of a molecule of interest, an in 
vitro ADCC assay, such as that described in US Patent No. 5,500,362 or 5,821,337 

30 may be performed. Useful effector cells for such assays include peripheral blood 



- 18- 



WO 01/74388 



PCT/US01/10382 



mononuclear cells (PBMC) and Natural Killer (NK) cells. Alternatively, or 
additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g., 
in a animal model such as that disclosed in Clynes et al. PNAS (USA) 95:652-656 
(1998). 

5 "Human effector cells" are leukocytes which express one or more FcRs and 

perform effector functions. Preferably, the cells express at least FcyRlH and carry out 
ADCC effector function. Examples of human leukocytes which mediate ADCC 
include peripheral blood mononuclear cells (PBMC), natural killer (NK) cells, 
monocytes, cytotoxic T cells and neutrophils; with PBMCs and NK cells being 
10 preferred. 

The terms "Fc receptor" or "FCR"are used to describe a receptor that binds to 
the Fc region of an antibody. 

The preferred FcR is a native sequence human FcR. Moreover, a preferred 
FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of 

15 the FcyRI, FcyRII, and Fey RIE subclasses, including allelic variants and alternatively 
spliced forms of these receptors. FcyRII receptors include FcyRJIA (an "activating 
receptor") and FcyREB (an "inhibiting receptor"), which have similar amino acid 
sequences that differ primarily in the cytoplasmic domains thereof. Activating 
receptor FcyRUA contains an immunoreceptor tyrosine-based activation motif 

20 (ITAM) in its cytoplasmic domain. Inhibiting receptor FcyRUB contains an 

immunoreceptor tyrosine-based inhibition motif (ITM) in its cytoplasmic domain, 
(see Daeron, Annu. Rev. Immunol. 15:203-234 (1997)). FcRs are reviewed in Ravetch 
and Kinet, Annu. Rev. Immunol 9:457-92 (1991); Capel et al., hnmunomethods 
4:25-34 (1994); and de Haas et al., J. Lab. Clin. Med. 126:330-41 (1995). Other FcRs, 

25 including those to be identified in the future, are encompassed by the term "FCR" 
herein. The term also includes the neonatal receptor, FcRn, which is responsible for 
the transfer of maternal IgGs to the fetus (Guyer et al., J. Immunol. 1 17:587 (1976) 
and Kim et al., J. Immunol. 24:249 (1994)). 

"Complement dependent cytotoxicity" or "CDC" refer to the ability of a 

30 molecule to lyse a target in the presence of complement. The complement activation 



- 19- 



WO 01/74388 PCT/US01/10382 

pathway is initiated by the binding of the first component of the complement system 
(Clq) to a molecule (e.g. an antibody) complexed with a cognate antigen. To assess 
complement activation, a CDC assay, e.g. as described in Gazzano-Santoro et al., J. 
Immunol. Methods 202: 1 63 (1 996), may be performed. 
5 "Growth inhibitory 1 ' antagonists are those which prevent or reduce 

proliferation of a cell expressing an antigen to which the antagonist binds. For 
example, the antagonist may prevent or reduce proliferation of B cells in vitro and/or 
in vivo. 

Antagonists which "induce apoptosis" are those which induce programmed 
1 0 cell death, e.g. of a B cell, as determined by standard apoptosis assays, such as binding 
of annexin V, fragmentation of DNA, cell shrinkage, dilation of endoplasmic 
reticulum, cell fragmentation, and/or formation of membrane vesicles (called 
apoptotic bodies). 

The term "antibody" herein is used in the broadest sense and specifically 
15 covers intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies 
(e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody 
fragments so long as they exhibit the desired biological activity. 

"Antibody fragments" comprise a portion of an intact antibody, preferably 
comprising the antigen-binding or variable region thereof. Examples of antibody 
20 fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies; 
single-chain antibody molecules; and multispecific antibodies formed from antibody 
fragments. 

"Native antibodies" are usually heterotetrameric glycoproteins of about 
150,000 daltons, composed of two identical light (L) chains and two identical heavy 

25 (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, 
while the number of disulfide linkages varies among the heavy chains of different 
immunoglobulin isotypes. Each heavy and light chain also has regularly spaced 
intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) 
followed by a number of constant domains. Each light chain has a variable domain at 

30 one end (VL) and a constant domain at its other end; the constant domain of the light 



-20- 



WO 01/74388 



PCT/US01/10382 



chain is aligned with the first constant domain of the heavy chain, and the light-chain 
variable domain is aligned with the variable domain of the heavy chain. Particular 
amino acid residues are believed to form an interface between the light chain and 
heavy chain variable domains. 
5 The term "variable" refers to the fact that certain portions of the variable 

domains differ extensively in sequence among antibodies and are used in the binding 
and specificity of each particular antibody for its particular antigen. However, the 
variability is not evenly distributed throughout the variable domains of antibodies. It 
is concentrated in three segments called hypervariable regions both in the light chain 

10 and the heavy chain variable domains. The more highly conserved portions of variable 
domains are called the framework regions (FRs). The variable domains of native 
heavy and light chains each comprise four FRs, largely adopting a P-sheet 
configuration, connected by three hypervariable regions, which form loops 
connecting, and in some cases forming part of, the (3 sheet structure. The 

1 5 hypervariable regions in each chain are held together in close proximity by the FRs 
and, with the hypervariable regions from the other chain, contribute to the formation 
of the antigen-binding site of antibodies (see Kabat et at, Sequences of Proteins of 
Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, 
Bethesda, MD. (1991)). The constant domains are not involved directly in binding an 

20 antibody to an antigen, but exhibit various effector functions, such as participation of 
the antibody in antibody dependent cellular cytotoxicity (ADCC). 

Papain digestion of antibodies produces two identical antigen-binding 
fragments, called "Fob" fragments, each with a single antigen-binding site, and a 
residual "Fc" fragment, whose name reflects its ability to crystallize readily. Pepsin 

25 treatment yields an F(ab'2 fragment that has two antigen-binding sites and is still 
capable of crosslinking antigen. 

"Fv" is the minimum antibody fragment which contains a complete 
antigen-recognition and antigen-binding site. This region consists of a dimer of one 
heavy chain and one light chain variable domain in tight, non-covalent association. It 

30 is in this configuration that the three hypervariable regions of each variable domain 



-21 - 



WO 01/74388 



PCT/US01/10382 



interact to define an antigen-binding site on the surface of the VH-VL dimer. 
Collectively, the six hypervariable regions confer antigen binding specificity to the 
antibody. However, even a single variable domain (or half of an Fv comprising only 
three hypervariable regions specific for an antigen) has the ability to recognize and 
5 bind antigen, although at a lower affinity than the entire binding site. 

The Fab fragment also contains the constant domain of the light chain and the 
first constant domain (CHI) of the heavy chain. Fab' fragments differ from Fab 
fragments by the addition of a few residues at the carboxy terminus of the heavy chain 
CHI domain including one or more cysteines from the antibody hinge region. Fab'-SH 

10 is the designation herein for Fab' in which the cysteine residue(s) of the constant 

domains bear at least one free thiol group. F(ab")Z antibody fragments originally were 
produced as pairs of Fab' fragments which have hinge cysteines between them. Other 
chemical couplings of antibody fragments are also known. 

The "light chains" of antibodies (immunoglobulins) from any vertebrate 

15 species can be assigned to one of two clearly distinct types, called kappa (x) and 
lambda (k), based on the amino acid sequences of their constant domains. 

Depending on the amino acid sequence of the constant domain of their heavy 
chains, antibodies can be assigned to different classes. There are five major classes of 
intact antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these maybe further 

20 divided into subclasses (isotypes), e.g., IgG 1, IgG2, IgG3, IgG4, IgA, and IgA2. The 
heavy-chain constant domains that correspond to the different classes of antibodies 
are called a, 8, s, y, and R, respectively. The subunit structures and three-dimensional 
configurations of different classes of immunoglobulins 
are well known. 

25 "Single-chain Fv" or "scFv" antibody fragments comprise the VH and VL 

domains of antibody, wherein these domains are present in a single polypeptide chain. 
Preferably, the Fv polypeptide further comprises a polypeptide linker between the VH 
and VL domains which enables the scFv to form the desired structure for antigen 
binding. For a review of scFv see Pluckthun in The Pharmacology of Monoclonal 

30 Antibodies, vol. 1 13, Rosenburg and Moore, eds., Springer- Verlag, New York, pp. 



-22- 



WO 01/74388 



PCT/US01/10382 



269-315 (1994). 

The term "diabodies" refers to small antibody fragments with two 
antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) 
connected to a light-chain variable domain (VL) in the same polypeptide chain (VH - 
5 VL). By using a linker that is too short to allow pairing between the two domains on 
the same chain, the domains are forced to pair with the complementary domains of 
another chain and create two antigen binding sites. Diabodies are described more fully 
in, for example, EP 404,097; WO 93/11161; and Hollinger et al, Proc. Nad. Acad. 
Sci. USA, 90:6444.-6448 (1993). 

10 The term "monoclonal antibody" as used herein refers to an antibody obtained 

from a population of substantially homogeneous antibodies, i.e., the individual 
antibodies comprising the population are identical except for possible naturally 
occurring mutations that may be present in minor amounts. Monoclonal antibodies are 
highly specific, being directed against a single antigenic site. Furthermore, in contrast 

15 to conventional (polyclonal) antibody preparations which typically include different 
antibodies directed against different determinants (epitopes), each monoclonal 
antibody is directed against a single determinant on the antigen. In addition to their 
specificity, the monoclonal antibodies are advantageous in that they are synthesized 
by the hybridoma culture, uncontaminated by other immunoglobulins. The modifier 

20 "monoclonal" indicates the character of the antibody as being obtained from a 
substantially homogeneous population of antibodies, and is not to be construed as 
requiring production of the antibody by any particular method. For example, the 
monoclonal antibodies to be used in accordance with the present invention may be 
made by the hybridoma method first described by Kohler et al, Nature, 256:495 

25 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Patent No. 
4,816,567). The "monoclonal antibodies" may also be isolated from phage antibody 
libraries using the techniques described in Clackson et al, Nature, 352:624-628 
(1991) and Marks et al, J. Mol. Biol, 222:581-597 (1991), for example. 

The monoclonal antibodies herein specifically include "chimeric" antibodies 

30 (immunoglobulins) in which a portion of the heavy and/or light chain is identical with 



-23- 



WO 01/74388 



PCT/US01/10382 



or homologous to corresponding sequences in antibodies derived from a particular 
species or belonging to a particular antibody class or subclass, while the remainder of 
the chain(s) is identical with or homologous to corresponding sequences in antibodies 
derived from another species or belonging to another antibody class or subclass, as 
5 well as fragments of such antibodies, so long as they exhibit the desired biological 
activity (U.S. Patent No. 4,816,567; Morrison et al, Proc. Natl. Acad. Sci. USA, 
81:6851-6855 (1984)). Chimeric antibodies of interest herein include "primatized" 
antibodies comprising variable domain antigen binding sequences derived from a 
non-human primate (e.g. Old World Monkey, such as baboon, rhesus or cynomolgus 

1 0 monkey) and human constant region sequences (US Pat No. 5,693,780). 

"Humanized" forms of non-human (e.g., murine) antibodies are chimeric 
antibodies that contain minimal sequence derived from non-human immunoglobulin. 
For the most part, humanized antibodies are human immunoglobulins (recipient 
antibody) in which residues from a hypervariable region of the recipient are replaced 

1 5 by residues from a hypervariable region of a non-human species (donor antibody) 
such as mouse, rat, rabbit or nonhuman primate having the desired specificity, 
affinity, and capacity. In some instances, framework region (FR) residues of the 
human immunoglobulin are replaced by corresponding non-human residues. 
Furthermore, humanized antibodies may comprise residues that are not found in the 

20 recipient antibody or in the donor antibody. These modifications are made to further 
refine antibody performance. In general, the humanized antibody will comprise 
substantially all of at least one, and typically two, variable domains, in which all or 
substantially all of the hypervariable loops correspond to those of a non-human 
immunoglobuhn and all or substantially all of the FRs are those of a human 

25 immunoglobulin sequence. The humanized antibody optionally also will comprise at 
least a portion of an immunoglobulin constant region (Fc), typically that of a human 
immunoglobulin. For further details, see Jones et al. Nature 321:522-525 (1986); 
Riechmann et al, Nature 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol. 
2:593-596 (1992). 

30 The term "hypervariable region" when used herein refers to the amino acid 



-24- 



WO 01/74388 PCT/US01/10382 

residues of an antibody which are responsible for antigen-binding. The hypervariable 
region comprises amino acid residues from a "complementarity determining region" 
or "CDR" (e.g. residues 24-34 (LI), 50-56 (L2) and 89-97 (L3) in the light chain 
variable domain and 3 1-35 (HI), 50-65 (H2) and 95-102 (H3) in the heavy chain 
5 variable domain; Kabat et al, Sequences of Proteins of Immunological Interest, 5th 
Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)) 
and/or those residues from a "hypervariable loop" (e.g. residues 26-32 (LI), 50-52 
(L2) and 91-96 (L3) in the light chain variable domain and 26-32 (HI), 53-55 (H2) 
and 96-101 (H3) in the heavy chain variable domain; Chothia and Lesk J. Mol. Biol. 

10 196:901-917 (1987)). "Framework" or "FR" residues are those variable domain 

residues other than the hypervariable region residues as herein defmed. An antagonist 
"which binds" an antigen of interest, e.g. a B cell surface marker, is one capable of 
binding that antigen with sufficient affinity and/or avidity such that the antagonist is 
useful as a therapeutic agent for targeting a cell expressing the antigen. 

15 Examples of antibodies which bind the CD20 antigen include: "C2B8" which 

is now called "rituximab" ("RITUXAN®") (US Patent No. 5,736,137, expressly 
incorporated herein by reference); the yttrium- [90] -labeled 2138 murine antibody 
designated "Y2B8" (US Patent No. 5,736,137, expressly incorporated herein by 
reference); murine IgG2a "131" optionally labeled with 1311 to generate the 

20 "131I-B1" antibody (BEXXARTM) (US Patent No. 5,595,721, expressly incorporated 
herein by reference); murine monoclonal antibody " 1F5" (Press et al. Blood 
69(2):584-591 (1987)); "chimeric 2H7" antibody (US Patent No. 5,677,180, expressly 
incorporated herein by reference); and monoclonal antibodies L27, G28-2, 93-1 133, 
B-Cl or NU-B2 available from the International Leukocyte Typing Workshop 

25 (V alentine et al, In: Leukocyte Typing III (McMichael, Ed., p. 440, Oxford University 
Press (1987)). Examples of antibodies which bind the CD 19 antigen include the 
anti-CD 19 antibodies in Hekman et al, Cancer Immunol. Immunother. 32:364-372 
(1991) and Vlasveld et al. Cancer Immunol. Immunother. 40:37-47(1995); and the B4 
antibody in Kiesel et al. Leukemia Research 11, 12: 1119 (1987). 

30 The terms "rituximab" or "RITUXAN®" herein refer to the genetically 



-25- 



WO 01/74388 PCT/US01/10382 

engineered chimeric murine/humati monoclonal antibody directed against the CD20 
antigen and designated "C2B8" in US Patent No. 5,736,137, expressly incorporated 
herein by reference. The antibody is an IgG, kappa irnmunoglobulin containing 
murine light and heavy chain variable region sequences and human constant region 
5 sequences. Rituximab has a binding affinity for the CD20 antigen of approximately 
8.0nM. 

An "isolated" antagonist is one which has been identified and separated and/or 
recovered from a component of its natural environment. Contaminant components of 
its natural environment are materials which would interfere with diagnostic or 

10 therapeutic uses for the antagonist, and may include enzymes, hormones, and other 
proteinaceous or nonproteinaceous solutes. In preferred embodiments, the antagonist 
will be purified (1) to greater than 95% by weight of antagonist as determined by the 
Lowry method, and most preferably more than 99% by weight, (2) to a degree 
sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence 

15 by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under 

reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain. 
Isolated antagonist includes the antagonist in situ within recombinant cells since at 
least one component of the antagonist's natural environment will not be present. 
Ordinarily, however, isolated antagonist will be prepared by at least one purification 

20 step. "Mammal" for purposes of treatment refers to any animal classified as a 
mammal, including humans, domestic and farm animals, and zoo, sports, or pet 
animals, such as dogs, horses, cats, cows, etc. Preferably, the mammal is human. 

"Treatment" refers to both therapeutic treatment and prophylactic or 
preventative measures. Those in need of treatment include those already with the 

25 disease or disorder as well as those in which the disease or disorder is to be prevented. 
Hence, the mammal may have been diagnosed as having the disease or disorder or 
may be predisposed or susceptible to the disease. 

The expression "therapeutically effective amount" refers to an amount of the 
antagonist which is effective for preventing, ameliorating or treating the autoimmune 

30 disease in question. The term "immunosuppressive agent" as used herein for adjunct 



-26- 



WO 01/74388 PCT/US01/10382 

therapy refers to substances that act to suppress or mask the immune system of the 
mammal being treated herein. This would include substances that suppress cytokine 
production, downregulate or suppress self-antigen expression, or mask the MHC 
antigens. 

5 Examples of such agents include 2-amino-6-aryl-5-substituted pyrirnidines 

(see U.S. Pat. No. 4,665,077, the disclosure of which is incorporated herein by 
reference); azathioprine; cyclophosphamide; bromocryptine; danazol; dapsone; 
glutaraldehyde (which masks the MHC antigens, as described in U.S. Pat. No. 
4,120,649); anti-idiotypic antibodies for MHC antigens and MHC fragments; 

10 cyclosporin A; steroids such as glucocorticosteroids, e.g., prednisone, 

methylprednisolone, and dexamethasone; cytokine or cytokine receptor antagonists 
including anti-interferon-y, -(3, or-a antibodies, anti-tumornecrosis factor-a 
antibodies, anti-tumornecrosis factor-(i antibodies, anti-mterleukin-2 antibodies and 
anti-IL-2 receptor antibodies; anti-LFA-1 antibodies, including anti-CD 11a and anti- 

15 CD 18 antibodies; anti-L3T4 antibodies; heterologous anti-lymphocyte globulin; pan-T 
antibodies, preferably antiCD3 or anti-CD4/CD4a antibodies; soluble peptide 
containing a LFA-3 binding domain (WO 90/08187 published 7/26/90); streptokinase; 
TGF-0; streptodornase; RNA or DNA from the host; FK506; RS-61443; 
deoxyspergualin; rapamycin; T-cell receptor (Cohen et al, U.S. Pat. No. 5,114,721); 

20 T-cell receptor fragments (Offher et al, Science 251 : 430-432 (1991); WO 90/1 1294; 
Ianeway, Nature, 341: 482 (1989); and WO 91/01133); and T cell receptor antibodies 
(EP 340,109) such as TLOB9. 

The term "cytotoxic agent" as used herein refers to a substance that inhibits or 
prevents the function of cells and/or causes destruction of cells. The term is intended 

25 to include radioactive isotopes (e.g. I 131 , Y 90 , Ar 211 , P 32 , Re 188 , Re 186 , Sm 153 , B 212 and 
others), chemotherapeutic agents, and toxins such as small molecule toxins or 
enzymatically active toxins of bacterial, fungal, plant or animal origin, or fragments 
thereof. 

A "chemotherapeutic agent" is a chemical compound useful in the treatment of 
30 cancer. Examples of chemotherapeutic agents include alkylating agents such as 



-27- 



WO 01/74388 PCT/US01/10382 

thiotepa and cyclosphosphamide (CYTOXANTM); alkyl sulfonates such as busulfan, 
improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, 
and uredopa; ethylenimines and methylamelamines including altretamine, 
triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and 
5 trimethylolomelamine; nitrogen mustards such as chlorambucil, chlornaphazine, 
cholophosphamide, estramustine, ifosfamide, mechlorethamine, mecMorethamine 
oxide hydrochloride, melphalan, novembiehin, phenesterine, prednimustine, 
trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, 
fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, 

10 actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, 

carabicin, carminomycin, carzinophilin, chromoinycins, dactinomycin, daunorubicin, 
detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, 
idambicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, 
olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, 

15 streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; 

anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues 
such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as 
fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as 
ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, 

20 doxifluridine, enocitabine, floxuridine, 5-FU; androgens such as calusterone, 

dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals 
such as aminoglutethimide, mitotane, trilostane; fohc acid replenisher such as frolinic 
acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; 
bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; 

25 elformthine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; 
lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; 
phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK®; 
razoxane; sizofrran; spirogermanium; tenuazonic acid; triaziquone; 2, 
2\2"-tricMorotriemylamine; urethan; vindesine; dacarbazine; mannomustine; 

30 mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); 



-28- 



WO 01/74388 



PCT/US01/10382 



cyclophosphamide; thiotepa; taxoids, e.g. pachtaxel (TAXOLO, Bristol-Myers Squibb 
Oncology, Princeton, NJ) and doxetaxel (TAXOTEW, Rh6ne-Poulenc Rorer, Antony, 
France); chlorambucil; gemcitabine; 6-tbioguanine; mercaptopurine; methotrexate; 
platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide 
5 (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; 
novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-11; 
topoisomerase inhibitor RFS 2000; difluoromemylorrdthine (DMFO); retinoic acid; 
esperamicins; capecitabine; and pharmaceutically acceptable salts, acids or derivatives 
of any of the above. Also included in this definition are anti-hormonal agents that act 

10 to regulate or inhibit hormone action on tumors such as anti-estrogens including for 
example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4 
hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene 
(Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, 
leuprohde, and goserelin; and pharmaceutically acceptable salts, acids or derivatives 

15 of any of the above. 

The term "cytokine" is a generic term for proteins released by one cell 
population which act on another cell as intercellular mediators. Examples of such 
cytokines are lymphokines,, monokines, and traditional polypeptide hormones. 
Included among the cytokines are growth hormone such as human growth hormone, 

20 N-methionyl human growth hormone, and bovine growth hormone; parathyroid 
hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones 
such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and 
luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin; 
placental lactogen; tumor necrosis factor-a and -0; mullerian-inhibiting substance; 

25 mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth 
factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF-P; platelet 
growth factor; transforming growth factors (TGFs) such as TGF-a and TGF-0; 
insulin-like growth factor-I and -H; erythropoietin (EPO); osteoinductive factors; 
interferons such as interferon-a, -P, and -y; colony stimulating factors (CSFs) such as 

30 macrophage-CSF (M-CSF); granulocyte-macrophage-CSF (GM-CSF); and 



-29- 



WO 01/74388 PCT/US01/10382 

granulocyte-CSF (GCSF); interleukins QLs) such as IL-1 , IL-la, IL-2, IL-3, IL-4, IL-5, 
JL-6, JL-7, JL-8, JL-9, IL-1 1, IL-12, EL-15; a tumor necrosis factor such as TNF-a or 
TNF-P; and other polypeptide factors including LIF and kit ligand (KL). As used 
herein, the term cytokine includes proteins from natural sources or from recombinant 
5 cell culture and biologically active equivalents of the native sequence cytokines. 
The term "prodrug" as used in this application refers to a precursor or 
derivative form of a pharmaceutically active substance that is less cytotoxic to tumor 
cells compared to the parent drug and is capable of being enzymatically activated or 
converted into the more active parent form. See, e.g., Wihnan, "Prodrugs in Cancer 

10 Chemotherapy" Biochemical Society Transactions, 14, pp. 375-382, 615th Meeting 
Belfast (1986) and Stella et al, "Prodrugs: A Chemical Approach to Targeted Drug 
Delivery," Directed Drug Delivery, Borchardt et al, (ed.), pp. 247-267, Humana Press 
(1985). The prodrugs of this invention include, but are not limited to, 
phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing 

1 5 prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated 
prodrugs, (3 -lactam-containing prodrugs, optionally substituted 
phenoxyacetamide-containing prodrugs or optionally substituted 
phenylacetamide-containing prodrugs, 5 fluorocytosine and other 5-fluorouridine 
prodrugs which can be converted into the more active cytotoxic free drag. Examples 

20 of cytotoxic drugs that can be derivatized into a prodrug form for use in this invention 
include, but are not limited to, those chemotherapeutic agents described above. 

A "liposome" is a small vesicle composed of various types of lipids, 
phospholipids and/or surfactant which is useful for delivery of a drug (such as the 
antagonists disclosed herein and, optionally, a chemotherapeutic agent) to a mammal. 

25 The components of the liposome are commonly arranged in a bilayer formation, 

similar to the lipid arrangement of biological membranes. The term "package insert" 
is used to refer to instructions customarily included in commercial packages of 
therapeutic products, that contain information about the indications, usage, dosage, 
administration, contraindications and/or warnings concerning the use of such 

30 therapeutic products. 



-30- 



WO 01/74388 PCT/US01/10382 



n. Production of Antagonists 

The methods and articles of manufacture of the present invention use, or 
incorporate, an antagonist which binds to a B cell surface marker and/or a cytokine. 
5 Accordingly, methods for generating such antagonists will be described here. The B 
cell surface marker or cytokine to be used for production of, or screening for, 
antagonists) may be, e.g., a soluble form of the antigen or a portion thereof, 
containing the desired epitope. Alternatively, or additionally, cells expressing the B 
cell surface marker at their cell surface can be used to generate, or screen for, 
10 antagonist(s). Other forms of the B cell surface marker useful for generating 

antagonists will be apparent to those skilled in the art. Preferably, the B cell surface 
marker is the CD 19 or CD20 antigen. Preferably, the cytokine is IL-10. 

While the preferred antagonist is an antibody, antagonists other man 
antibodies are contemplated herein. For example, the antagonist may comprise a 
1 5 small molecule antagonist optionally fused to, or conjugated with, a cytotoxic agent 
(such as those described herein). Libraries of small molecules may be screened against 
the B cell surface marker of interest herein in order to identify a small molecule which 
binds to that antigen. The small molecule may further be screened for its antagonistic 
properties and/or conjugated with a cytotoxic agent. 
20 The antagonist may also be a peptide generated by rational design or by phage 

display (see, e.g., W098/35036 published 13 August 1998). In one embodiment, the 
molecule of choice may be a "CDR mimic" or antibody analogue designed based on 
the CDRs of an antibody. While such peptides may be antagonistic by themselves, the 
peptide may optionally be fused to a cytotoxic agent so as to add or enhance 
25 antagonistic properties of the peptide. 

A description follows as to exemplary techniques for the production of the 
antibody antagonists used in accordance with the present invention. 
Polyclonal antibodies 
Polyclonal antibodies are preferably raised in animals by multiple 
30 subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an 



-31 - 



WO 01/74388 PCT/USO 1/10382 

adjuvant. It may be useful to conjugate the relevant antigen to a protein that is 
immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin, serum 
albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional or 
derivatizing agent, for example, maleimidobenzoyl sulfosuccinimide ester 
5 (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine 
residues), glutaraldehyde, succinic anhydride, SOC12, or R1N=C=NR, where R and 
RI are different alkyl groups. Animals are immunized against the antigen, 
immunogenic conjugates, or derivatives by combining, e.g., 100 pg or 5 wg of the 
protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund's 

10 complete adjuvant and injecting the solution intradermally at multiple sites. One 

month later the animals are boosted with 1 15 to 1/10 the original amount of peptide or 
conjugate in Freund's complete adjuvant by subcutaneous injection at multiple sites. 
Seven to 14 days later the animals are bled and the serum is assayed for antibody titer. 
Animals are boosted until the titer plateaus. Preferably, the animal is boosted with the 

15 conjugate of the same antigen, but conjugated to a different protein and/or through a 
different cross-linking reagent. Conjugates also can be made in recombinant cell 
culture as protein fusions. Also, aggregating agents such as alum are suitably used to 
enhance the immune response. 

(it) Monoclonal antibodies 

20 Monoclonal antibodies are obtained from a population of substantially 

homogeneous antibodies, Le., the individual antibodies comprising the population are 
identical except for possible naturally occurring mutations that may be present in 
minor amounts. Thus, the modifier "monoclonal" indicates the character of the 
antibody as not being a mixture of discrete antibodies. For example, the monoclonal 

25 antibodies may be made using the hybridoma method first described by Kohler et al, 
Nature, 256:495 (1975), or may be made by recombinant DNA methods (U.S. Patent 
No. 4,816,567). 

In the hybridoma method, a mouse or other appropriate host animal, such as a 
hamster, is immunized as hereinabove described to elicit lymphocytes that produce or 
30 are capable of producing antibodies that will specifically bind to the protein used for 



-32- 



WO 01/74388 PCT/US01/10382 

immunization. Alternatively, lymphocytes maybe immunized in vitro. Lymphocytes 
then are fused with myeloma cells using a suitable fusing agent, such as polyethylene 
glycol, to form a hybridoma 

cell [Goding, Monoclonal Antibodies: Principles and Practice, pp.59-103 (Academic 
5 Press, 1986)]. 

The hybridoma cells thus prepared are seeded and grown in a suitable culture 
medium that preferably contains one or more substances that inhibit the growth or 
survival of the unfused, parental myeloma cells. For example, if the parental myeloma 
cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or 

1 0 HPRT), the culture medium for the hybridomas typically will include hypoxanthine, 
arninopterin, and thymidine (HAT medium), which substances prevent the growth of 
HGPRT-deficient cells. 

Preferred myeloma cells are those that fuse efficiently, support stable 
high-level production of antibody by the selected antibody-producing cells, and are 

15 sensitive to a medium such as HAT medium. Among these, preferred myeloma cell 
lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-1 1 
mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, 
California USA, and SP-2 or X63-Ag8-653 cells available from the American Type 
Culture Collection, Rockville, Maryland USA. Human myeloma and mouse human 

20 heteromyeloma cell lines also have been described for the production of human 
monoclonal antibodies [Kozbor, J. Immunol, 133:3001 (1984); Brodeur et al, 
Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel 
Dekker, Inc., New York, 1987)]. 

Culture medium in which hybridoma cells are growing is assayed for 

25 production of monoclonal antibodies directed against the antigen. Preferably, the 
binding specificity of monoclonal antibodies produced by hybridoma cells is 
determined by immunoprecipitation or by an in vitro binding assay, such as 
radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). The 
binding affinity of the monoclonal antibody can, for example, be determined by the 

30 Scatchard analysis of Munson et al, Anal Biochem., 107:220 (1980). 



-33- 



WO 01/74388 



PCT/US01/10382 



After hybridoma cells are identified that produce antibodies of the desired 
specificity, affinity, and/or activity, the clones maybe subcloned by limiting dilution 
procedures and grown by standard methods (Goding, Monoclonal Antibodies: 
Principles and Practice, pp.59-103 (Academic Press, 1986)). Suitable culture media 
5 for this purpose include, for example, D-MEM or RPMI-1640 medium. In addition, 
the hybridoma cells may be grown in vivo as ascites tumors in an animal. 

The monoclonal antibodies secreted by the subclones are suitably separated 
from the culture medium, ascites fluid, or serum by conventional immunoglobulin 
purification procedures such as, for example, protein A-Sepharose, hydroxylapatite 

10 chromatography, gel electrophoresis, dialysis, or affinity chromatography. 

DNA encoding the monoclonal antibodies is readily isolated and sequenced 
using conventional procedures (e.g., by using oligonucleotide probes that are capable 
of binding specifically to genes encoding the heavy and light chains of murine 
antibodies). The hybridoma cells serve as a preferred source of such DNA. Once 

1 5 isolated, the DNA may be placed into expression vectors, which are then transfected 
into host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) 
cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to 
obtain the synthesis of monoclonal antibodies in the recombinant host cells. Review 
articles on recombinant expression in bacteria of DNA encoding the antibody include 

20 Skerra et ah, Curr. Opinion in Immunol., 5:256-262 (1993) and Phickthun, Immunol. 
Revs., 130:151-188 (1992). 

In a further embodiment, antibodies or antibody fragments can be isolated 
from antibody phage libraries generated using the techniques described in McCafferty 
et al, Nature, 348:552-554 (1990). Clackson et al, Nature, 352:624-628 (1991) and 

25 Marks et al, J. Mol. Biol, 222:581-597 (1991) describe the isolation of murine and 
human antibodies, respectively, using phage libraries. Subsequent publications 
describe the production of high affinity (nM range) human antibodies by chain 
shuffling (Marks et al, BiolTechnology, 10:779-783 (1992)), as well as combinatorial 
infection and in vivo recombination as a strategy for constructing very large phage 

30 libraries (Waterhouse et al, Nuc. Acids. Res., 21 :2265-2266 (1 993)). Thus, these 



-34- 



WO 01/74388 PCTYUS01/10382 

techniques are viable alternatives to traditional monoclonal antibody hybridoma 
techniques for isolation of monoclonal antibodies. 

The DNA also may be modified, for example, by substituting the coding 
sequence for human heavy- and light-chain constant domains in place of the 
5 homologous murine sequences (U.S. Patent No. 4,8 1 6,567; Morrison, et al, Proc. 
Natl Acad. Sci. USA, 81:6851 (1984)), or by covalently joining to the 
immunoglobulin coding sequence all or part of the coding sequence for a 
non-immunoglobuUn polypeptide. Typically such non-immunoglobulin polypeptides 
are substituted for the constant domains of an antibody, or they are substituted for the 
10 variable domains of one anu^en-combining site of an antibody to create a chimeric 
bivalent antibody comprising one antigen-combining site having specificity for an 
antigen and another antigen combining site having specificity for a different antigen. 

(Hi) Humanized antibodies 

Methods for humanizing non-human antibodies have been described in the art. 

1 5 Preferably, a humanized antibody has one or more amino acid residues introduced into 
it from a source which is non-human. These non-human amino acid residues are often 
referred to as "import" residues, which are typically taken from an "import" variable 
domain. Humanization can be essentially performed following the method of Winter 
and co-workers (Jones et al, Nature, 321:522-525 (1986); Riechmann et al, Nature, 

20 332:323-327 (1988); Verhoeyen et aL, Science, 239:1534-1536 (1988)), by 

substituting hypervariable region sequences for the corresponding sequences of a 
human antibody. Accordingly, such "humanized" antibodies are chimeric antibodies 
(U.S. Patent No. 4,816,567) wherein substantially less than an intact human variable 
domain has been substituted by the corresponding sequence from a non-human 

25 species. In practice, humanized antibodies are typically human antibodies in which 
some hypervariable region residues and possibly some FR residues are substituted by 
residues from analogous sites in rodent antibodies. 

The choice of human variable domains, both fight and heavy, to be used in 
making the humanized antibodies is very important to reduce antigenicity. According 

30 to the so-called "best-fit" method, the sequence of the variable domain of a rodent 



-35- 



WO 01/74388 



PCT/US01/10382 



antibody is screened against the entire library of known human variable-domain 
sequences. The human sequence which is closest to that of the rodent is then accepted 
as the human framework region (FR) for the humanized antibody (Sims et al, J. 
Immunol, 151:2296 (1993); Chothia et al, J. Mol. Biol, 196:901 (1987)). Another 
5 method uses a particular framework region derived from the consensus sequence of all 
human antibodies of a particular subgroup of light or heavy chains. The same 
framework may be used for several different humanized antibodies (Carter et aL, 
Proc. Nad. Acad. Sci. USA, 89:4285 (1992); Presta et al, J. Immunol, 151:2623 
(1993)). 

10 It is further important that antibodies be humanized with retention of high 

affinity for the antigen and other favorable biological properties. To achieve this goal, 
according to a preferred method, humanized antibodies are prepared by a process of 
analysis of the parental sequences and various conceptual humanized products using 
three dimensional models of the parental and humanized sequences. 

15 Three-dimensional immunoglobulin models are commonly available and are familiar 
to those skilled in the art. Computer programs are available which illustrate and 
display probable three-dimensional conformational structures of selected candidate 
immunoglobulin sequences. Inspection of these displays permits analysis of the likely 
role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., 

20 the analysis of residues that influence the ability of the candidate immunoglobulin to 
bind its antigen, hi Ibis way, FR residues can be selected and combined from the 
recipient and import sequences so that the desired antibody characteristic, such as 
increased affinity for the target antigen(s), is achieved. In general, the hypervariable 
region residues are directly and most substantially involved in influencing antigen 

25 binding. 

(iv) Human antibodies 
As an alternative to humanization, human antibodies can be generated. For 
example, it is now possible to produce transgenic animals (e.g., mice) that are 
capable, upon immunization, of producing a full repertoire of human antibodies in the 
30 absence of endogenous immunoglobulin production. For example, it has been 



-36- 



WO 01/74388 



PCT/US01/10382 



described that the homozygous deletion of the antibody heavy-chain joining region 
(JH) gene in chimeric and germ-line mutant mice results in complete inhibition of 
endogenous antibody production. Transfer of the human germ-line immunoglobulin 
gene array in such germ-line mutant mice will result in the production of human 
5 antibodies upon antigen challenge. See, e.g., Jakobovits et al, Proc. Mad. Acad. Set 
USA, 90:2551 (1993); Jakobovits et al, Nature, 362:255-258 (1993); Bruggermann et 
al, Year inlmmuno., 7:33 (1993); and US Patent Nos. 5,591,669, 5,589,369 and 
5,545,807. Alternatively, phage display technology (McCafferty etal, Nature 
348:552-553 (1990)) can be used to produce human antibodies and antibody 

10 fragments in vitro, from immunoglobulin variable (V) domain gene repertoires from 
unimmunized donors. According to this technique, antibody V domain genes are 
cloned in-frame into either a major or minor coat protein gene of a filamentous 
bacteriophage, such as Ml 3 or fd, and displayed as functional antibody fragments oh 
the surface of the phage particle. Because the filamentous particle contains a 

1 5 single-stranded DNA copy of the phage genome, selections based on the functional 
properties of the antibody also result in selection of the gene encoding the antibody 
exhibiting those properties. Thus, the phage mimics some of the properties of the B 
cell. Phage display can be performed in a variety of formats; for their review see, e.g. 
Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology 

20 3:564-571(1993). Several sources of V-gene segments can be used for phage display. 
Clackson et al, Nature, 352: 624-628 (1991) isolated a diverse array of 
anti-oxazolone antibodies from a small random combinatorial library of V genes 
derived from the spleens of immunized mice. A repertoire of V genes from 
uinmmunized human donors can be constructed and antibodies to a diverse array of 

25 antigens (including self-antigens) can be isolated essentially following the techniques 
described by Marks et al, J. Mol Biol. 222:581-597 (1991), or Griffith et al, EMBO 
J. 12:725-734 (1993). See, also, US Patent Nos. 5,565,332 and 5,573,905. Human 
antibodies may also be generated by in vitro activated B cells (see US Patents 
5,567,610 and 5,229,275). 

30 (v) Antibody fragments 



-37- 



WO 01/74388 PCT/US01/10382 



Various techniques have been developed for the production of antibody 
fragments. Traditionally, these fragments were derived via proteolytic digestion of 
intact antibodies (see, e.g., Morimoto et al., Journal of Biochemical and Biophysical 
Methods 24:107-1 17 (1992) and Brennan et al, Science, 229:81 (1985)). However, 
5 these fragments can now be produced directly by recombinant host cells. For example, 
the antibody fragments can be isolated from the antibody phage libraries discussed 
above. Alternatively, Fab'-Sli fragments can be directly recovered from E. coli and 
chemically coupled to form F(ab')2 fragments [Carter et al, Bio/Technology 
10:163-167 (1992)]. According to another approach, F(ab')2 fragments can be isolated 

10 directly from recombinant host cell culture. Other techniques for the production of 
antibody fragments will be apparent to the skilled practitioner. In other embodiments, 
the antibody of choice is a single chain Fv fragment (scFv). See WO 93/16185; US 
Patent No. 5,571,894; and US Patent No. 5,587,458. The antibody fragment may also 
be a "linear antibody", e.g., as described in US Patent 5,641,870 for example. Such 

1 5 linear antibody fragments may be monospecific or bispecific. 
(vi) Bispecific antibodies 
Bispecific antibodies are antibodies that have binding specificities for at least 
two different epitopes. Exemplary bispecific antibodies may bind to two different 
epitopes of the B cell surface marker. Other such antibodies may bind a first B cell 

20 marker and further bind a second B cell surface marker. Alternatively, an anti-B cell 
marker binding arm may be combined with an arm which binds to a triggering 
molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2 or CD3), or Fc 
receptors for IgG (FcyR), such as FcyPJ (CD64), FcyPJI (CD32) and FcyRm (CD 16) 
so as to focus cellular defense mechanisms to the B cell. Bispecific antibodies may 

25 also be used to localize cytotoxic agents to the B cell. These antibodies possess a B 
cell marker-binding arm and an arm which binds the cytotoxic agent (e.g. saporin, 
anti-interferon-a, vinca alkaloid, ricin A chain, methotrexate or radioactive isotope 
hapten). Bispecific antibodies can be prepared as full length antibodies or antibody 
fragments (e.g. F(ab')Z bispecific antibodies). 

30 Methods for making bispecific antibodies are known in the art. Traditional 



-38- 



WO 01/74388 



PCT/US01/10382 



production of full length bispecific antibodies is based on the coexpression of two 
immunoglobulin heavy chain-light chain pairs, where the two chains have different 
specificities (Millstein et al., Nature, 305:537-539 (1983)). Because of the random 
assortment of immunoglobulin heavy and light chains , these hybridomas (quadromas) 
5 produce a potential mixture of 1 0 different antibody molecules, of which only one has 
the correct bispecific structure. Purification of the correct molecule, which is usually 
done by affinity chromatography steps, is rather cumbersome, and the product yields 
are low. Similar procedures are disclosed in WO 93/08829, and in Traunecker et al, 
EMBOJ, 10:3655-3659 (1991). 

10 According to a different approach, antibody variable domains with the desired 

binding specificities (antibody-antigen combining sites) are fused to immunoglobulin 
constant domain sequences. The fusion preferably is with an immunoglobulin heavy 
chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It 
is preferred to have the first heavy-chain constant region (CHI) containing the site 

1 5 necessary for light chain binding, present in at least one of the fusions. DNAs 
encoding the immunoglobulin heavy chain fusions and, if desired, the 
immunoglobulin light chain, are inserted into separate expression vectors, and are 
co-transfected into a suitable host organism. This provides for great flexibility in 
adjusting the mutual proportions of the three polypeptide fragments in embodiments 

20 when unequal ratios of the three polypeptide chains used in the construction provide 
the optimum yields. It is, however, possible to insert the coding sequences for two or 
all three polypeptide chains in one expression vector when the expression of at least 
two polypeptide chains in equal ratios results in high yields or when the ratios are of 
no particular significance. 

25 m a preferred embodiment of this approach, the bispecific antibodies are 

composed of a hybrid immunoglobulin heavy chain with a first binding specificity in 
one arm, and a hybrid immunoglobulin heavy chain light chain pair (providing a 
second binding specificity) in the other arm. It was found that this asymmetric 
structure facilitates the separation of the desired bispecific compound from unwanted 

30 immunoglobulin chain combinations, as the presence of an immunoglobulin light 



-39- 



WO 01/74388 



PCT/US01/10382 



chain in only one half of the bispecific molecule provides for a facile way of 
separation. This approach is disclosed in WO 94/04690. For further details of 
generating bispecific antibodies see, for example, Suresh et al, Methods in 
Enzymology, 121:210(1986). 
5 According to another approach described in US Patent No. 5,731,168, the 

interface between a pair of antibody molecules can be engineered to maximize the 
percentage of heterodimers which are recovered from recombinant cell culture. The 
preferred interface comprises at least a part of the CH3 domain of an antibody 
constant domain. In this method, one or more small amino acid side chains from the 

1 0 interface of the first antibody molecule are replaced with larger side chains (e.g. 
tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the 
large side chain(s) are created on the interface of the second antibody molecule by 
replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). 
This provides' a mechanism for increasing the yield of the heterodimer over other 

15 unwanted end-products such as homodimers. 

Bispecific antibodies include cross-linked or "heteroconjugate" antibodies. For 
example, one of the antibodies in the heteroconjugate can be coupled to avidin, the 
other to biotin. Such antibodies have, for example, been proposed to target immune 
system cells to unwanted cells (US Patent No. 4,676,980), and for treatment of HIV 

20 infection (WO 91/00360, WO 92/200373, and EP 03089). Heteroconjugate antibodies 
may be made using any convenient cross-linking methods. Suitable cross-linking 
agents are well known in the art, and are disclosed in US Patent No. 4,676,980, along 
with a number of cross-linking techniques. 

Techniques for .generating bispecific antibodies from antibody fragments have 

25 also been described in the literature. For example, bispecific antibodies can be 

prepared using chemical linkage. Brennan et al, Science, 229:81 (1985) describe a 
procedure wherein intact antibodies are proteolytically cleaved to generate F(ab*)2 
fragments. These fragments are reduced in the presence of the dithiol complexing 
agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide 

30 formation. The Fab' fragments generated are then converted to thionitrobenzoate 

-40- 



WO 01/74388 



PCT/US01/10382 



(TNB) derivatives. One of the Fab'-TNB derivatives is then reconverted to the 
Fab'-thiol by reduction with mercaptoethylamine and is mixed with an equirnolar 
amount of the other Fab'-TNB derivative, to form the bispecific antibody. The 
bispecific antibodies produced can be used as agents for the selective immobilization 
5 of enzymes. 

Recent progress has facilitated the direct recovery of Fab'-SH fragments from 
E. coli, which can be chemically coupled to form bispecific antibodies. Shalaby et al, 
J. Exp. Med., 175: 217-225 (1992) describe the production of a fully humanized 
bispecific antibody F(ab')2 molecule. Each Fab' fragment was separately secreted from 

10 E. coli and subjected to directed chemical coupling in vitro to form the bispecific 
antibody. The bispecific antibody thus formed was able to bind to cells 
overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the 
lytic activity of human cytotoxic lymphocytes against human breast tumor targets. 
Various techniques for making and isolating bispecific antibody fragments 

1 5 directly from recombinant cell culture have also been described. For example, 

bispecific antibodies have been produced using leucine zippers. Kostelny et al, J. 
Immunol, 148(5): 1547-1 553 (1992). The leucine zipper peptides from the Fos and 
Jun proteins were linked to the Fab' portions of two different antibodies by gene 
fusion. The antibody homodimers were reduced at the hinge region to form monomers 

20 and then re-oxidized to form the antibody heterodimers. This method can also be 
utilized for the production of antibody homodimers. The "diabody" technology 
described by Hollinger et al, Proc. Natl Acad. Sci. USA, 90:6444-6448 (1993) has 
provided an alternative mechanism for making bispecific antibody fragments. The 
fragments comprise a heavy-chain variable domain (VH) connected to a light-chain 

25 variable domain (VL) by a linker which is too short to allow pairing between the two 
domains on the same chain. 

Accordingly, the VH arid VL domains of one fragment are forced to pair with 
the complementary VL and VH domains of another fragment, thereby forming two 
antigen-binding sites. Another strategy for making bispecific antibody fragments by 

30 the use of single-chain Fv (sFv) dimers has also been reported. See Gruber et al, J. 



-41 - 



WO 01/74388 PCTYUS01/10382 

Immunol, 152:5368 (1994). Antibodies with more than two valencies are 
contemplated. For example, trispecific antibodies can be prepared. Tutt et al. J. 
Immunol. 147: 60 (1991). 

m. Conjugates and Other Modifications of the Antagonist 
5 The antagonists used in the methods or included in the articles of manufacture 

herein are optionally conjugated to a cytotoxic agent. Chemotherapeutic agents useful 
in the generation of such antagonist-cytotoxic agent conjugates have been described 
above. 

Conjugates of an antagonist and one or more small molecule toxins, such as a 

10 cahcheamicin, a maytansine (US Patent No. 5,208,020), a trichothene, and CC1065 
are also contemplated herein. In one embodiment of the invention, the antagonist is 
conjugated to one or more maytansine molecules (e.g. about 1 to about 10 
maytansinemolecules per antagonist molecule). Maytansine may, for example, be 
converted to May-SS-Me which may be reduced to May-SH3 and reacted with 

15 modified antagonist (Chari et al. Cancer Research 52: 127-131 (1992)) to generate a 
maytansinoid-antagonist conjugate. 

Alternatively, the antagonist is conjugated to one or more calicheamicin 
molecules. The calicheamicin family of antibiotics are capable of producing 
double-stranded DNA breaks at sub-picomolar concentrations. Structural analogues 

20 of calicheamicin which may be used include, but are not limited to, 'yJ 1 , a2 1 , a3 1 , 
N-acetyl-yl", PSAG and 01 1 (Hinman et al. Cancer Research 53: 3336-3342 (1993) 
and Lode et al. Cancer Research 58: 2925-2928 (1998)). 

Enzymatically active toxins and fragments thereofwhich can be used include 
diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain 

25 (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, 

alpha-sarcin, 41euritesfordii proteins, dianthin proteins, Phytolaca americana proteins 
(PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria 
officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin and the 
tricothecenes. See, for example, WO 93/21232 published October 28, 1993. 

30 The present invention further contemplates antagonist conjugated with a 



-42- 



WO 01/74388 PCTAJS01/10382 

compound with nucleolytic activity (e.g. a ribonuclease or a DNA endonuclease such 
as a deoxyribonuclease; DNase). A variety of radioactive isotopes are available for 
the production of radioconjugated antagonists. Examples include Ate",113',1125, Y9o 
Re 186, Re 188, Sml53, Bi212 P32 and radioactive isotopes of Lu. Conjugates of the 
5 antagonist and cytotoxic agent may be made using a variety of bifunctional protein 
coupling agents such as N-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), 
succinimidyl-4-(N-maleimidomethyl) cyclohexane-l-carboxylate, iminothiolane (IT), 
bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active 
esters (such as disuccinimidyl suberate), aidehydes (such as glutareldehyde), bis azido 

10 compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives 
(such as bis-(pdiazoniumbenzoyl)-emylenediamine), diisocyanates (such as tolyene 
2,6-diisocyanate), and bis-active fluorine compounds (such as 
l,5-difluoro-2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared 
as described in Vitetta et al. Science 238:1098 (1987). Carbon- 14-labeled 

15 l-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is 
an exemplary chelating agent for conjugation of radionucleotide to the antagonist. See 
W094/1 1026. The linker may be a "cleavable linker" facilitating release of the 
cytotoxic drug in the cell. For example, an acid-labile linker, peptidase-sensitive 
linker, dimethyl linker or msmfide-containing linker (Chari et ah Cancer Research 

20 52: 127-13 1 (1992)) may be used. Alternatively, a fusion protein comprising the 
antagonist and cytotoxic agent may be made, e.g. by recombinant techniques or 
peptide synthesis. 

In yet another embodiment, the antagonist may be conjugated to a "receptor" 
(such streptavidin) for utilization in tumor pretargeting wherein the 

25 antagonist-receptor conjugate is administered to the patient, followed by removal of 
unbound conjugate from the circulation using a clearing agent and then administration 
of a "ligand" (e.g. avidin) which is conjugated to a cytotoxic agent (e.g. a 
radionucleotide). The antagonists of the present invention may also be conjugated 
with a prodrug-activating enzyme which converts a prodrug (e.g. a pepndyl 

30 chemotherapeutic agent, see W081/01 145) to an active anti-cancer drug. See, for 



-43- 



WO 01/74388 



PCTYUS01/10382 



example, WO 88/07378 and U.S. Patent No. 4,975,278. 

The enzyme component of such conjugates includes any enzyme capable of 
acting on a prodrug in such a way so as to covert it into its more active, cytotoxic 
form. Enzymes that are useful in the method of this invention include, but are not 
5 limited to, alkaline phosphatase useful for converting phosphate-cOntaining prodrugs 
into free drugs; arylsulfatase useful for converting sulfate containing prodrugs into 
free drugs; cytosine deaminase useful for converting non-toxic 5-fluorocytosine into 
the anti-cancer drug, 5-fluorouracil; proteases, such as serratia protease, thermolysin, 
subtilisin, carboxypeptidases and cathepsins (such as cathepsins B and L), that are 

10 useful for converting peptide-containing prodrugs into free drugs; 

D-alanylcarboxypeptidases, useful for converting prodrugs that contain D-amino acid 
substituents; carbohydrate cleaving enzymes such as li-galactosidase and 
neuraminidase useful for converting glycosylated prodrugs into free drugs; 
(3-lactamase useful for converting drugs derivatized with (3-lactams into free drugs; 

1 5 and penicillin amidases, such as penicillin V amidase or penicillin G amidase, useful 
for converting drugs derivatized at their amine nitrogens with phenoxyacetyl or 
phenylacetyl groups, respectively, into free drugs. Alternatively, antibodies with 
enzymatic activity, also known in the art as "abzymes", can be used to convert the 
prodrugs of the invention into free active drugs (see, e.g., Massey, Nature 328: 

20 457-458 (1987)). Antagonist-abzyme conjugates can be prepared as described herein 
for delivery of the abzyme to a tumor cell population. 

The enzymes of this invention can be covalently bound to the antagonist by 
techniques well known in the art such as the use of the heterobifunctional crosslinking 
reagents discussed above. Alternatively, fusion proteins comprising at least the 

25 antigen binding region of an antagonist of the invention linked to at least a 

functionally active portion of an enzyme of the invention can be constructed using 
recombinant DNA techniques well known in the art [see, e.g., Neuberger et al, 
Nature, 312: 604-608 (1984)]. 

Other modifications of the antagonist are contemplated herein. For example, 

30 the antagonist may be linked to one of a variety of nonproteinaceous polymers, e.g., 



-44- 



WO 01/74388 



PCT/US01/10382 



polyethylene glycol, polypropylene glycol, polyoxyalkylenes, or copolymers of 
polyethylene glycol and polypropylene glycol. The antagonists disclosed herein may 
also be formulated as liposomes. Liposomes containing the antagonist are prepared by 
methods known in the art, such as described in Epstein et al, Proc. Mad. Acad Sci. 

5 USA, 82:3688 (1985); Hwang et al, Proc. Natl Acad. Sci. USA, 77:4030 (1980); U.S. 
Pat. Nos. 4,485,045 and 4,544,545; and W097/38731 published October 23, 1997. 
Liposomes with enhanced circulation time are disclosed in U.S. Patent No. 5,013,556. 

Particularly useful liposomes can be generated by the reverse phase 
evaporation method with a lipid composition comprising phosphatidylcholine, 

0 cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are 
extruded through filters of defined pore size to yield liposomes with the desired 
diameter. Fab' fragments of an antibody of the present invention can be conjugated to 
the liposomes as described in Martin et al, J. Biol. Chem. 257: 286-288 (1982) via a 
disulfide interchange reaction. A chemotherapeutic agent is optionally contained 

5 within the liposome. See Gabizon et al. J. National Cancer Inst.81(19)1484 (1989). 
Amino acid sequence modifications) of protein or peptide antagonists described 
herein are contemplated. For example, it may be desirable to improve the binding 
affinity and/or other biological properties of the antagonist. 

Amino acid sequence variants of the antagonist are prepared by introducing 

0 appropriate nucleotide changes into the antagonist nucleic acid, or by peptide 

synthesis. Such modifications include, for example, deletions from, and/or insertions 
into and/or substitutions of, residues within the amino acid sequences of the 
antagonist. Any combination of deletion, insertion, and substitution is made to arrive 
at the final construct, provided that the final construct possesses the desired 

5 characteristics. The amino acid changes also may alter post-translational processes of 
the antagonist, such as changing the number or position of glycosylation sites. 

A useful method for identification of certain residues or regions of the 
antagonist that are preferred locations for mutagenesis is called "alanine scanning 
mutagenesis" as described by Cunningham and Wells Science, 244:1081-1085 (1989). 

0 Here, a residue or group of target residues are identified (e.g., charged residues such 



-45- 



WO 01/74388 



PCT/US01/10382 



as arg, asp, his, lys, and glu) and replaced by a neutral or negatively charged amino 
acid (most preferably alanine or polyalanine) to affect the interaction of the amino 
acids with antigen. Those amino acid locations demonstrating functional 
sensitivity to the substitutions then are refined by introducing further or other variants 
5 at, or for, the sites of substitution. Thus, while the site for introducing an amino acid 
sequence variation is predetermined, the nature of the mutation per se need not be 
predeterrnined. For example, to analyze the performance of a mutation at a given site, 
ala scanning or random mutagenesis is conducted at the target codon or region and the 
expressed antagonist variants are screened for the desired activity. 

10 Amino acid sequence insertions include amino- and/or carboxyl-terminal 

fusions ranging in length from one residue to polypeptides containing a hundred or 
more residues, as well as intrasequence insertions of single or multiple amino acid 
residues. Examples of terminal insertions include an antagonist with an N-terminal 
methionyl residue or the antagonist fused to a cytotoxic polypeptide. Other insertional 

1 5 variants of the antagonist molecule include the fusion to the N- or C-terminus of the 
antagonist of an enzyme, or a polypeptide which increases the serum half-life of the 
antagonist. 

Another type of variant is an amino acid substitution variant. These variants 
have at least one amino acid residue in the antagonist molecule replaced by different 
20 residue. The sites of greatest interest for substitutional mutagenesis of antibody 
antagonists include the hypervariable regions, but FR alterations are also 
contemplated. 

Conservative substitutions are shown in Table 1 under the heading of 
"preferred substitutions". If such substitutions result in a change in biological activity, 
25 then more substantial changes, denominated "exemplary substitutions" in Table 1 , or 
as further described below in reference to arnino acid classes, may be introduced and 
the products screened. 



Table 1 



Original 


Exemplary 


Preferred 


Residue 


Substitutions 


Substitutions 



-46- 



WO 01/74388 



PCT/US01/10382 



Ala (A) 


val; leu; ile 


val 


Arg(R) 


lys; gin; asn 


lys 


Asn(N) 


gin; his; asp, lys; arg 


gin 


Asp(D) 


glu; asn 


glu 


Cys(C) 


ser; ala 


ser 


Gin CO) 


asn; glu 


asn 


Glu (E) 


asp; gin 


asp 


Gly(G) 


ala 


ala 


His (H) 


asn; gin; lys; arg 


arg 


He (I) 


leu; val; met; ala; 
phe; norleucine 


ICU 


Lea (L) 


norleucine; ile; val; 
met; ala; phe 


ile 


Lys(K) 


arg; gin; asn 


arg 


Met(M) 


leu; phe; ile 


leu 


Phe(F) 


leu; val; ile; ala; tyr 


tyr 


Pro(P) 


ala 


ala 


Ser (S) 


thr 


thr 


Thr(T) 


ser 


ser 


TIP (W) 


tyr; phe 


tyr 


Tyr(Y) 


trp; phe; thr; ser 


phe 


Val(V) 


ile; leu; met; phe; 
ala; norleucine 


ICU 



Substantial modifications in the biological properties of the antagonist are 
accomplished by selecting substitutions that differ significantly in their effect on 
mamtaining (a) the structure of the polypeptide backbone in the area of the 
5 substitution, for example, as a sheet or helical conformation, (b) the charge or 
hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. 
Naturally occurring residues are divided into groups based on common side-chain 
properties: 

(1) hydrophobic: norleucine, met, ala, val, leu, ile; 
10 (2) neutral hydrophilic: cys, ser, thr; 

(3) acidic: asp, glu; 

(4) basic: asn, gin, his, lys, arg; 

(5) residues that influence chain orientation: gly, pro; and 

(6) aromatic: trp, tyr, phe. 



-47- 



WO 01/74388 



PCT/US01/10382 



Non-conservative substitutions will entail exchanging a member of one of 
these classes for another class. 

Any cysteine residue not involved in maintaining the proper conformation of 
the antagonist also may be substituted, generally with serine, to improve the oxidative 
5 stability of the molecule and prevent aberrant crosslinking. Conversely, cysteine 

bond(s) may be added to the antagonist to improve its stability (particularly where the 
antagonist is an antibody fragment such as an Fv fragment). 

A particularly preferred type of substitutional variant involves substituting one 
or more hypervariable region residues of a parent antibody. Generally, the resulting 

10 variant(s) selected for further development will have improved biological properties 
relative to the parent antibody from which they are generated. A convenient way for 
generating such substitutional variants is affinity maturation using phage display. 
Briefly, several hypervariable region sites (e.g.6-7 sites) are mutated to generate all 
possible amino substitutions at each site. The antibody variants thus generated are 

1 5 displayed in a monovalent fashion from filamentous phage particles as fusions to the 
gene m product of Ml 3 packaged within each particle. The phage-displayed variants 
are then screened for their biological activity (e.g. binding affinity) as herein 
disclosed. In order to identify candidate hypervariable region sites for modification, 
alanine scanning mutagenesis can be performed to identify hypervariable region 

20 residues contributing significantly to antigen binding. Alternatively, or in additionally, 
it may be beneficial to analyze a crystal structure of the antigen-antibody complex to 
identify contact points between the antibody and antigen. Such contact residues and 
neighboring residues are candidates for substitution according to the techniques 
elaborated herein. Once such variants are generated, the panel of variants is subjected 

25 to screening as described herein and antibodies with superior properties in one or 
more relevant assays may be selected for further development. 

Another type of amino acid variant of the antagonist alters the original 
glycosylation pattern of the antagonist. By altering is meant deleting one or more 
carbohydrate moieties found in the antagonist, and/or adding one or more 

30 glycosylation sites that are not present in the antagonist. 



-48- 



WO 01/74388 PCT/US01/10382 

Glycosylation of polypeptides is typically either N-linked or O-linked. 
N-linked refers to the attachment of the carbohydrate moiety to the side chain of an 
asparagine residue. The tripeptide sequences asparagine-X-serine and 
asparagine-X-threonine, where X is any amino acid except proline, are the recognition 
5 sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side 
chain. Thus, the presence of either of these tripeptide sequences in a polypeptide 
creates a potential glycosylation site. O-linked glycosylation refers to the attachment 
of one of the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino 
acid, most commonly serine or threonine, although 5-hydroxyproline or 

10 5-hydroxylysine may also be used. Addition of glycosylation sites to the antagonist is 
conveniently accomplished by altering the amino acid sequence such that it contains 
one or more of the above-described tripeptide sequences (for N-linked glycosylation 
sites). The alteration may also be made by the addition of, or substitution by, one or 
more serine or threonine residues to the sequence of the original antagonist (for 

15 O-linked glycosylation sites). 

Nucleic acid molecules encoding amino acid sequence variants of the 
antagonist are prepared by a variety of methods known in the art. These methods 
include, but are not limited to, isolation from a natural source (in the case of naturally 
occurring amino acid sequence variants) or preparation by oligonucleotide-mediated 

20 (or site directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an 
earlier prepared variant or a non-variant version of the antagonist. 

It may be desirable to modify the antagonist of the invention with tespect to 
effector function, e.g. so as to enhance antigen-dependent cell-mediated cyotoxicity 
(ADCC) and/or complement dependent cytotoxicity (CDC) of the antagonist. This 

25 may be achieved by introducing one or more amino acid substitutions in an Fc region 
of an antibody antagonist. Alternatively or additionally, cysteine residue(s) may be 
introduced in the Fc region, thereby allowing interchain disulfide bond formation in 
this region. The homodimeric antibody thus generated may have improved 
internalization capability and/or increased complement-mediated cell killing and 

30 antibody-dependent cellular cytotoxicity (ADCC). See Caron et al, J. Exp Med. 



-49- 



WO 01/74388 PCT/US01/10382 

176:1191-1195 (1992) and Shopes, B. J. Immunol. 148:2918-2922 (1992). 
Homodimeric antibodies with enhanced anti-tumor activity may also be prepared 
using heterobifunetional cross-linkers as described in Wolff et al. Cancer Research 
53:2560-2565 (1993). Alternatively, an antibody can be engineered which has dual Fc 
5 regions and may thereby have enhanced complement lysis and 

ADCC capabilities. See Stevenson et al Anti-Cancer Drug Design 3:219-230 (1989). 

To increase the serum half life of the antagonist, one may incorporate a 
salvage receptor binding epitope into the antagonist (especially an antibody fragment) 
as described in US Patent 5,739,277, for example. As used herein, the term "salvage 
10 receptor binding epitope" refers to an epitope of the Fc region of an IgG molecule 
(e.g., IgGl, IgG2, IgG3, or IgG4) that is responsible for increasing the in vivo serum 
half-life of the IgG molecule. 

IV. Pharmaceutical Formulations 

Therapeutic formulations of the antagonists used in accordance with the 

1 5 present invention are prepared for storage by mixing an antagonist or antagonists 

having the desired degree of purity with optional pharmaceutically acceptable carriers, 
excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. 
Ed. (1980)), in the form of lyophilized formulations or aqueous solutions. Acceptable 
carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and 

20 concentrations employed, and include buffers such as phosphate, citrate, and other 
organic acids; antioxidants including ascorbic acid and methionine; preservatives 
(such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; 
benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl 
parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 

25 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) 
polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; 
hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, 
glulamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, 
and other carbohydrates including glucose, mannose, or dextrins; chelating agents 

30 such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt forming 



-50- 



WO 01/74388 PCT/US01/10382 

counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or 
non-ionic surfactants such as TWEENTM, PLURONICSTM or polyethylene glycol 
(PEG). 

Exemplary anti-CD20 antibody formulations are described in W098/56418, 
5 expressly incorporated herein by reference. This publication describes a liquid 
multidose formulation comprising 40 mg/mL rituximab, 25 mM acetate, 150 mM 
trehalose, 0.9% benzyl alcohol, 0.02% polysorbate 20 at pH 5.0 that has a minimum 
shelf life of two years storage at 2-8 °C. Another anti-CD20 formulation of interest 
comprises 1 Omg/mL rituximab in 9.0 mg/mL sodium chloride, 7.35 mg/mL sodium 

10 citrate dihydrate, 0.7mg/mL polysorbate 80, and Sterile Water for Injection, pH 6.5. 
Lyophilized formulations adapted for subcutaneous administration are described in 
W097/04801. Such lyophilized formulations may be reconstituted with a suitable 
diluent to a high protein concentration and the reconstituted formulation may be 
administered subcutaneously to the mammal to be treated herein. 

1 5 The formulation herein may also contain more than one active compound zi.; 

necessary for the particular indication being treated, preferably those with 
complementary activities that do not adversely affect each other. For example, it may 
be desirable to further provide a cytotoxic agent, chemotherapeutic agent, cytokine or 
immunosuppressive agent (e.g. one which acts on T cells, such as cyclosporin or an 

20 antibody that binds T cells, e.g. one which binds LFA-1). The effective amount of 
such other agents depends on the amount of antagonist present in the formulation, the 
type of disease or disorder or treatment, and other factors discussed above. These are 
generally used in the same dosages and with a<iministration routes as used 
hereinbefore or about from 1 to 99% of the heretofore employed dosages. 

25 The active ingredients may also be entrapped in microcapsules prepared, for 

example, by coacervation techniques or by interfacial polymerization, for example, 
hydroxymethylcellulose or gelatin-microcapsules and poly(methylmethacylate) 
microcapsules, respectively, in colloidal drug delivery systems (for example, 
liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) 

30 or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical 



-51- 



WO 01/74388 



PCT/US01/10382 



Sciences 1 6th edition, Osol, A. Ed. (1 980). 

Sustained-release preparations maybe prepared. Suitable examples of 
sustained-release preparations include semipermeable matrices of solid hydrophobic 
polymers containing the antagonist, which matrices are in the form of shaped articles, 
5 e.g. films, or microcapsules. Examples of sustained-release matrices include 
polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or 
poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic 
acid and y ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic 
acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable 
10 microspheres composed of lactic acid glycolic acid copolymer and leuprolide acetate), 
and poly-D-(-)-3-hydroxybutyric acid. 

The formulations to be used for in vivo administration must be sterile. This is 
readily accomplished by filtration through sterile filtration membranes. 

15 V. Treatment with the Antagonist 

A composition comprising an antagonist which binds to a B cell surface 
antigen and a composition which contains a cytokine antagonist, e.g. an antibody, 
wherein both may be in the same composition will be formulated, dosed, and 
administered in a fashion consistent with good medical practice. Preferably, the anti- 

20 cytokine will comprise an anti-ILlO antibody and the B cell antagonist will comprise a 
B cell depleting antibody, preferably an anti-CD20 antibody such as Rituxan®. 
Factors for consideration in this context include the particular disease or disorder 
being treated, the particular mammal being treated, the clinical condition of the 
individual patient, the cause of the disease or disorder, the site of delivery of the 

25 agent, the method of administration, the scheduling of administration, and other 
factors known to medical practitioners. The therapeutically effective amount of the 
antagonist to be administered will be governed by such considerations. 

As a general proposition, the therapeutically effective amount of the antagonist 
administered parenterally per dose will be in the range of about 0.1 to 20 mg/kg of 

30 patient body weight per day, with the typical initial range of antagonist used being in 



-52- 



WO 01/74388 



PCT/US01/10382 



the range of about 2 to 10 mg/kg. 

The preferred antagonist is an antibody, e.g. an antibody such as RITUXAN®, 
which is not conjugated to a cytotoxic agent. Suitable dosages for an unconjugated 
antibody are, for example, in the range from about 20mg/mz to about I OOOmg/m2. 
5 In one embodiment, the dosage of the antibody differs from that presently 

recommended for RITUXAN®. For example, one may administer to the patient one 
or more doses of substantially less than 375mg/m2 of the antibody, e.g. where the 
dose is in the range from about 20mg/mz to about 250mg/m2, for example from about 
50mg/m2 to about 200mg/m2. 

10 Moreover, one may administer one or more initial dose(s) of the antibody 

followed by one or more subsequent dose(s), wherein the mg/m2 dose of the antibody 
in the subsequent dose(s) exceeds the mg/m2 dose of the antibody in the initial 
dose(s). For example, the initial dose may be in the range from about 20mg/m2 to 
about 250mg/m2 (e.g. from about 50mg/m2 to about 200mg/mz) and the subsequent 

15 dose may be in the range from about 250mg/m2 to about 1000mg/m2. 

As noted above, however, these suggested amounts of antagonist are subject to 
a great deal of therapeutic discretion. The key factor in selecting an appropriate dose 
and scheduling is the result obtained, as indicated above. 

For example, relatively higher doses may be needed initially for the treatment 

20 of ongoing and acute diseases. To obtain the most efficacious results, depending on 
the disease or disorder, the antagonist is administered as close to the first sign, 
diagnosis, appearance, or occurrence of the disease or disorder as possible or during 
remissions of the disease or disorder. 

The antagonist is administered by any suitable means, including parenteral, 

25 subcutaneous, intraperitoneal, intrapulmonary, and intranasal, and, if desired for local 
immunosuppressive treatment, intralesional administration. Parenteral infusions 
include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous 
administration. 

In addition, the antagonist may suitably be administered by pulse infusion, 
30 e.g., with declining doses of the antagonist. Preferably the dosing is given by 



-53- 



WO 01/74388 



PCT/US01/10382 



injections, most preferably intravenous or subcutaneous injections, depending in part 
on whether the administration is brief or chronic. 

One may administer other compounds, such as cytotoxic agents, 
chemotherapeutic agents, immunosuppressive agents and/or cytokines with the 
5 antagonists herein. The combined administration includes coadministration, using 
separate formulations or a single pharmaceutical formulation, and consecutive 
administration in either order, wherein preferably there is a time period while both (or 
all) active agents simultaneously exert their biological activities. 

Aside from administration of protein antagonists to the patient the present ■ 

1 0 application contemplates administration of antagonists by gene therapy. Such 
administration of nucleic acid encoding the antagonist is encompassed by the 
expression "administering a therapeutically effective amount of an antagonist". See, 
for example, W096/07321 published March 14, 1996 concerning the use of gene 
therapy to generate intracellular antibodies. 

1 5 There are two major approaches to getting the nucleic acid (optionally 

contained in a vector) into the patient's cells; in vivo and ex vivo. For in vivo delivery 
the nucleic acid is injected directly into the patient, usually at the site where the 
antagonist is required. For ex vivo treatment, the patient's cells are removed, the 
nucleic acid is introduced into these isolated cells and the modified cells are 

20 adrninistered to the patient either directly or, for example, encapsulated within porous 
membranes which are implanted into the patient (see, e.g. U.S. Patent Nos. 4,892,538 
and 5,283,187). There are a variety of techniques available for introducing nucleic 
acids into viable cells. The techniques vary depending upon whether the nucleic acid 
is transferred into cultured cells in vitro, or in vivo in the cells of the intended host. 

25 Techniques suitable for the transfer of nucleic acid into mammalian cells in vitro 
include the use of liposomes, electroporation, microinjection, cell fusion, 
DEAE-dextran, the calcium phosphate precipitation method, etc. A commonly used 
vector for ex vivo delivery of the gene is a retrovirus. 

The currently preferred in vivo nucleic acid transfer techniques include 

30 transfection with viral vectors (such as adenovirus, Herpes simplex I virus, or 



-54- 



WO 01/74388 PCT/US01/10382 

J 

adeno-associated virus) and lipid-based systems (useful lipids for lipid mediated 
transfer of the gene are DOTMA, DOPE and DC-Choi, for example). In some 
situations it is desirable to provide the nucleic acid source with an agent that targets 
the target cells, such as an antibody specific for a cell surface membrane protein or the 
5 target cell, a ligand for a receptor on the target cell, etc. Where liposomes are 
employed, proteins which bind to a cell surface membrane protein associated with 
endocytosis may be used for targeting and/or to facilitate uptake, e.g. capsid proteins 
or fragments thereof tropic for a particular cell type, antibodies for proteins which 
undergo internalization in cycling, and proteins that target intracellular localization 

10 and enhance intracellular half-life. The technique of receptor-mediated endocytosis is 
described, for example, by Wu et aL, J. Biol. Chem. 262:4429-4432 (1987); and 
Wagner et aL, Proc. Nad. Acad. Sci. USA 87:3410-3414 (1990). For review of the 
currently known gene marking and gene therapy protocols see Anderson et aL, 
Science 256:808-813 (1992). See also WO 93/25673 and the references cited therein. 

15 VI. Articles of Manufacture 

In another embodiment of the invention, an article of manufacture containing 
materials useful for the treatment of the diseases or disorders described above is 
provided. The article of manufacture comprises a container and a label or package 
insert on or associated with the container. Suitable containers include, for example, 

20 bottles, vials, syringes, etc. The containers maybe formed from a variety of materials 
such as glass or plastic. The container holds or contains a composition which is 
effective for treating the disease or disorder of choice and may have a sterile access 
port (for example the container may be an intravenous solution bag or a vial having a 
stopper pierceable by a hypodermic injection needle). At least one active agent in the 

25 composition is the antagonist which binds a B cell surface marker. Preferably CD20, 
and an anti-cytokine antibody, e.g. an anti-ILlO antibody. The label or package insert 
indicates that the composition is used for treating a patient having or predisposed to 
an autoimmune disease, such as those listed herein. The article of manufacture may 
further comprise a second container comprising a pharmaceutically- acceptable 

30 diluent buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered 



-55 - 



WO 01/74388 



PCT/US01/10382 



saline, Ringer's solution and dextrose solution. It may further include other materials 
desirable from a commercial and user standpoint, including other buffers, diluents, 
filters, needles, and syringes. 

Further details of the invention are illustrated by the following non-limiting 
5 Examples. The disclosure of all citations in the specification are expressly 
incorporated by reference. 

. Examples 

10 Example 1 

Treatment of Non-Hodgkin's Lymphoma 

A patient with non-Hodgkin's lymphoma is intravenously administered an 
anti-DLlO antibody at a dosage of 50mg/m 2 IV weekly for four weeks. Thereafter, the 
patient is administered RITUXAN® intravenously according to the following dosage 
15 schedules: 

(A) 50mg/m 2 IV day 1 

1 50mg/m 2 IV days on 8, 1 5 & 22 

(B) 150mg/m 2 rVdayl 

20 375mg/m 2 IV on days 8, 15 & 22 

(C) 375mg/m 2 IV on days 1, 8, 15 & 22 

This same patient is administered CHOP chemotherapy according to the 
regimen described in US Patent 5,736,137. 
25 After treatment, the patient is monitored to evaluate the effect on lymphoma 

status, e.g., number and size of tumors. 

Example 2 

Treatment of Solid Tumor in Advanced Stage 

30 A patient having an advanced colorectal cancer characterized by B cell 

involvement is treated concurrently with an anti-ILlO antibody and RITUXAN® at 
the same dosages as in Example 1 . 



-56- 



WO 01/74388 



PCT/US01/10382 



After treatment the patient is evaluated to determine whether such treatment 
has resulted in an anti-tumor response, e.g., based on tumor shrinkage, lower tumor 
antigen expression or other means of evaluating disease prognosis. 



-57- 



WO 01/74388 



PCT/US01/10382 



WHAT IS CLAIMED : 

1 . A method of avoiding, decreasing or overcoming the resistance of 
hematologic malignant cells or solid non-hematologic tumor cells to at least one 
chemotherapeutic agent, comprising administering an anti-cytokine antibody or 

5 fragment thereof or cytokine antagonist to a patient diagnosed with a hematologic 
malignancy or a solid, non-hematologic tumor prior, concurrent or after 
administration of at least one chemotherapeutic agent. 

2. The method of Claim 1, wherein said hematologic cells comprise B 
10 cell lymphoma or leukemia cells. 



3 . The method of Claim 2, wherein said B cell lymphoma is selected from 
the group consisting of low grade/ follicular non-Hodgkin' s lymphoma (NHL), small 
lymphocytic (SL) NHL, intermediate grade/ follicular NHL, intermediate grade 

15 diffuse NHL, high grade immunoblastic NHL, high grade lymphoblastic NHL, high 
grade small non-cleaved cell NHL, bulky disease NHL and Waldenstrom's 
Macroglobulinemia. 

4. The method of Claim 3, wherein said B cell lymphoma is low grade/ 
20 follicular non-Hodgkins lymphoma (NHL). 



5. The method of Claim 2, wherein said leukemic cell is selected from the 
group consisting of acute lymphoblastic leukemia, acute myelogenous leukemia, 
chronic lymphocytic leukemia, chronic myelogenous leukemia, lymphoblastic 

25 leukemia, lymphocytic leukemia, monocytic leukemia, myelogenous leukemia, and 
promyelocyte leukemia. 

6. The method of Claim 1, wherein said at least one chemotherapeutic 
agent is selected from the group consisting of CHOP, ICE, Mitozantrone, Cytarabine, 

30 DVP, ATRA, Idarubicin, hoelzer chemotherapy regime, La La chemotherapy regime, 



-58- 



WO 01/74388 



PCT/US01/10382 



ABVD, CEOP, 2-CdA, FLAG & IDA with or without subsequent G-CSF treatment), 
VAD, M & P, C-Weekly, ABCM, MOPP, DHAP, daunorubicin, doxorubicin, 
tamoxifen, toremifene, methotrexate, and cisplatin. 

5 7. The method of Claim 1, wherein said cytokine is selected from the 

group consisting of JL2, IL6, IL10 and TNF-alpha. 

8. The method of Claim 7, wherein said cytokine is IL10. 

10 9. The method of Claim 8, wherein said anti-ELl 0 antibody is a 

humanized or human monoclonal antibody. 

10. The method of Claim 9, wherein said anti-lLl 0 antibody is 
administered at a dosage of 0.01 to 1000 mg/kg body weight. 

15 

1 1 . The method of Claim 1 0, wherein the dosage of antibody ranges from 
about 0. 1 to 50 mg/kg of body weight. 

12. The method of Claim 1, wherein said anti-cytokine antibody is 
20 administered concurrently with and/or prior to said chemotherapeutic agent. 

13. The method of Claim 12, wherein said anti-cytokine antibody is 
administered concurrently or from about one hour to thirty days prior to 
administration of the chemotherapeutic agent. 

25 

14. The method of Claim 1, wherein the serum of said lymphoma patient is 
tested for cytokine profiles prior to administration of said anti-cytokine antibody or 
fragment thereof or antagonist. 



-59- 



WO 01/74388 



PCT/US01/10382 



15. A kit for administering the antibody or antagonist according to the 
method of Claim 1. 

16. A kit for testing cytokine profile according to the method of Claim 14. 

5 

17. A kit for testing cytokine profile and administering antibody or 
antagonist according to the method of Claim 16. 

18. A method of avoiding, decreasing or overcoming the resistance of 

10 hematologic malignant cells to a therapeutic agent, comprising administering an anti- 
cytokine antibody or cytokine antagonist to a patient diagnosed with a hematologic 
malignancy. 

19. A method of avoiding, decreasing or overcoming the resistance of 

1 5 hematologic malignant cells to apoptosis induced by a therapeutic agent, comprising 
administering an anti-cytokine antibody or cytokine antagonist to a patient diagnosed 
with a hematologic malignancy. 

20. The method of Claim 1 8, wherein said malignancy is a B cell 
20 lymphoma or leukemia. 

21. The method of Claim 19, wherein said malignancy is a B cell 
lymphoma or leukemia. 

25 22. A method of treating a patient with a hematologic malignancy who has 

relapsed following chemotherapy, comprising administering an anti-cytokine antibody 
or fragment thereof or cytokine antagonist to said patient. 



-60- 



WO 01/74388 PCT/US01/10382 



23. A method of treating a patient having a hematologic malignancy who 
is refractory to chemotherapy, comprising adrriinistering an anti-cytokine antibody or 
fragment thereof or cytokine antagonist to said patient. 

5 24. The method of Claim 21 , wherein said malignancy is a B cell 

lymphoma or leukemia. 

25 . A method of treating a patient with a hematologic malignancy who has 
relapsed following therapy with a therapeutic antibody or fragment, comprising 

10 administering an anti-cytokine antibody or fragment or cytokine antagonist to said 
patient. 

26. The method of Claim 23, wherein said therapeutic antibody is an anti- 
CD20, anti-CD 19, anti-CD22, anti-CD37, anti-CD40, or anti-CD28 antibody. 

15 

27 . The method of treating a patient with a hematologic malignancy who is 
refractory to therapy with a therapeutic antibody, comprising administering an anti- 
cytokine antibody or fragment or cytokine antagonist to said patient. 

20 28 . The method of Claim 25 , wherein said hematologic malignancy is B 

cell lymphoma or leukemia. 

29. A method of treating a B cell lymphoma patient comprising 
administering to said patient a therapeutically effective amount of a B cell depleting 

25 antibody simultaneously with or consecutively with in either order an anti-cytokine 
antibody or fragment. 

30. The method of Claim 29 wherein such B cell depleting antibody binds 
a B cell antigen selected from the group consisting of CD19, CD20, CD22, CD23, 



-61 - 



WO 01/74388 



PCT/US01/10382 



CD27, CD37, CD53, CD72, CD73, CD74, CD©78, CD79a, CD79b, CD80, CD81, 
CD82, CD83, CDw84, CD85 and CD86. 

3 1 . The method of Claim 2 1 wherein said B cell depleting antibody binds 

5 CD20. 

32. The method of Claim 29 wherein said B cell depleting antibody binds 

CD22. 

10 33. The method of Claim 29, further comprising adrninistration of at least 

one chemotherapeutic agent. 

34. The method of Claim 33, wherein said at least one chemotherapeutic 
agent is selected from the group consisting of CHOP, ICE, Mitozantrone, Cytarabine, 
1 5 DVP, ATRA, Idarubicin, hoelzer chemotherapy regime, La La chemotherapy regime, 
ABVD, CEOP, 2-CdA, FLAG & IDA with or without subsequent G-CSF treatment), 
VAD, M & P, C- Weekly, ABCM, MOPP, DHAP, daunorubicin, doxorubicin, 
methotrexate, and cisplatin. 

20 35. The method of Claim 33, wherein said anti-cytokine antibody or 

antagonist is administered prior to said anti-CD20 antibody and said at least one 
chemotherapeutic agent. 

36. The method of Claim 29, wherein said anti-cytokine antibody or 
25 antagonist is administered prior to said anti-CD20 antibody. 

37. The method of Claim 29, wherein said cytokine is selected from the 
group consisting of EL2, IL6, IL10 and TNF-alpha. 

30 38. The method of Claim 3 7, wherein said cytokine is IL10. 



-62- 



WO 01/74388 



PCT/US01/10382 



39. The method of Claim 29, wherein said anti-CD20 antibody is a 
chimeric, humanized or human anti-CD20 antibody. 

5 40. The method Claim of 39, wherein said anti-CD20 antibody is a 

chimeric anti-CD20 antibody. 

41 . The method of Claim 40, where said chimeric anti-CD20 antibody is 
Rituximab®. 

10 

42. The method of Claim 41, wherein said Rituximab® is administered at 
a dosage of 0.4 to 20 mg/kg body weight. 

43. The method of Claim 33, wherein said at least one chemotherapeutic 
1 5 agent is part of a CHOP chemotherapeutic regimen. 

44. The method of Claim 29, wherein said B cell lymphoma is selected 
from the group consisting of low grade/ follicular non-Hodgkin's lymphoma (NHL), 
small lymphocytic (SL) NHL, intermediate grade/ follicular NHL, intermediate grade 

20 diffuse NHL, high grade immunoblastic NHL, high grade lymphoblastic NHL, high 
grade small non-cleaved cell NHL, bulky disease NHL and Waldenstrom's 
Macroglobulinemia. 

45. The method of Claim 44, wherein said B cell lymphoma is non- 
25 Hodgkin's lymphoma (NHL). 

46. The method of Claim 45, wherein said B cell lymphoma is low-grade, 
follicular NHL. 



-63- 



WO 01/74388 PCT/US01/10382 



47. The method of Claim 29, wherein the serum of said lymphoma patient 
is tested for cytokine profiles prior to administration of said anti-cytokine antibody or 
antagonist. 

5 48. A kit for administering the anti-CD20 antibody and the anti-cytokine 

antibody or antagonist according to the method of Claim 29. 

49. A kit for testing cytokine profile and adrninistering anti-CD20 antibody 
and anti-cytokine antibody or antagonist according to the method of Claim 42. 

10 

50. A method for treating a tumor having B cell involvement comprising 
administering to a patient in need of such treatment an effective amount of an 
antibody specific to a cytokine and a B cell depleting antibody which binds to an 
antigen expressed by B cells. 

15 

51. A kit for administering the anti-CD20 antibody and the anti-cytokine 
antibody or antagonist according to the method of Claim 50. 

52. The method of Claim 50 wherein said anti-cytokine antibody binds to a 
20 cytokine selected from the group consisting of an interferon, interleukin, tumor 

necrosis factor, and colony stimulating factor. 

53 . The method of Claim 5 1 wherein said anti-cytokine antibody binds to a 
cytokine selected from the group consisting of an interferon, interleukin, tumor 

25 necrosis factor, and colony stimulating factor. 

54. The method of Claim 50 wherein said anti-cytokine antibody 
specifically binds IL-10. 



-64- 



WO 01/74388 



PCT/US01/10382 



55. The method of Claim 5 1 wherein said anti-cytokine antibody 
specifically binds IL- 10. 

56. The method of Claim 50 wherein the B cell antigen is selected from the 
5 group consisting of CD19, CD20, CD22, CD23, CD27, CD37, CD53, CD72, CD73, 

CD74, CD©78, CD79a, CD79b, CD80, CD81, CD82, CD83, CDw84, CD85 and 
CD86. 

57. A kit for administering the anti-CD20 antibody and the anti-cytokine 
10 antibody or antagonist according to the method of Claim 51. 

58. The method of Claim 56 wherein said B cell antigen is CD20. 

59. The method of Claim 58 wherein the anti-CD20 antibody is a human, 
15 humanized or chimeric anti-CD20 antibody. 

60. The method of Claim 59 wherein said antibody possesses ADCC 
and/or CDC activity. 

20 61 . The method of Claim 59 wherein said anti-CD20 induces apoptosis of 

B cells. 

62. The method of Claim 59 wherein said anti-CD20 antibody is 
Rituxan®, a chimeric anti-CD20 antibody produced by ATCC 691 19. 

25 

63 . The method of Claim 5 1 wherein said patient comprises a solid non- 
lymphoid tumor associated with a cancer selected from the group consisting of liver 
cancer, head and neck cancer, breast cancer, prostate cancer, testicular cancer, ovarian 
cancer, lung cancer, esophageal cancer, tracheal cancer, kidney cancer, bladder cancer, 

30 and colorectal cancer. 



-65- 



WO 01/74388 



PCT/US01/10382 



64. The method of Claim 50 wherein said B cell lymphoma is selected 
from the group consisting of low grade/ follicular non-Hodgkin's lymphoma (NHL), 
small lymphocytic (SL) NHL, intermediate grade/ follicular NHL, intermediate grade 
diffuse NHL, high grade immunoblastic NHL, high grade lymphoblastic NHL, high 

5 grade small non-cleaved cell NHL, bulky disease NHL and Waldenstrom' s 
Macroglobulinemia. 

65. The method of Claim 50 wherein said antibodies are administered by 
intravenous, intramuscular, intratumoral or intraperitoneal administration. 

10 

66. The method of Claim 51 wherein said antibodies are administered by 
intravenous, intramuscular, intratumoral or intraperitoneal administration. 

67. The method of Claim 5 1 when said solid tumor comprises a precancer, 
15 early stage (Stage I or II solid cancer), advanced cancer (after Stage II cancer) or 

metastasized cancer. 

68 . The method of Claim 5 1 wherein said patient has colorectal cancer or 
lung cancer. 

20 

69. A method of treating colorectal cancer or lung cancer having B cell 
involvement comprising administering to a patient in need of such treatment an 
effective amount of an antibody specific to DL-1 0 and a depleting anti-CD20 antibody. 

25 70. The method of Claim 69 wherein said depleting anti-CD20 antibody is 

a human, humanized or chimeric antibody. 

7 1 . The method of Claim 70 wherein said antibody is Rituxan® produced 
by ATCC 69119. 

30 



-66- 



WO 01/74388 



PCT/US01/10382 



72. A method of treating B cell lymphoma in a patient in need of such 
treatment which treatment includes the administration of an anti-ILlO antibody. 

73 . A method of treating non-Hodgkin' s lymphoma in a patient in need of 
5 such treatment which method comprises the administration of at least one anti-ILl 0 

antibody. 

74. A method of treating B cell lymphoma in a patient in need of such 
treatment which method includes the administration of anti-ILl 0 antibody and at least 

10 one B cell depleting antibody. 

75 . The method of Claim 63 wherein said B cell depleting antibody binds 
to a B cell antigen from the group consisting of CD 19, CD20, CD22, CD23, CD27, 
CD37, CD53, CD72, CD73, CD74, CDco78, CD79a, CD79b, CD80, CD81, CD82, 

15 CD83, CDw84, CD85 and CD86. 

76. A method of treating B cell lymphoma in a patient in need of such 
treatment which method comprises the administration of an anti-ILl 0 antibody and a 
B cell depleting anti-CD20 or anti-CD22 antibody. 

20 

77. A method of treating B cell lymphoma in a patient in need of such 
treatment comprising the administration of an anti-ILl 0 antibody and a B cell 
depleting anti-CD20 antibody. 

25 78 . A method of treating non-Hodgkin' s lymphoma in a patient in need of 

such treatment comprising the administration of an anti-ILl 0 antibody and a B cell 
depleting antibody. 



-67- 



WO 01/74388 



PCT/US01/10382 



79. A method of treating non-Hodgkin' s lymphoma in a patient in need of 
such treatment comprising the administration of an anti-ILl 0 antibody and a B cell 
depleting anti-CD20 antibody. 

5 80. A method of treating non-Hodgkin's lymphoma in a patient in need of 

such treatment comprising the aciministration of an anti-ILl 0 antibody and a B cell 
depleting anti-CD22 antibody. 

8 1 . The method of Claim 77 wherein said antibody is Rituxan® . 

10 

82. The method of Claim 79 wherein said antibody is Rituxan®. 

83 . A combination therapy for treating B cell lymphoma in a patient 
comprising the administration of a therapeutically effective amount of an anti-ILl 0 

15 antibody, a B cell depleting anti-CD20 antibody and chemotherapy. 

84. The method of Claim 83 wherein said anti CD20 antibody is 
Rituxan®. 

20 85. The method of Claim 83 wherein said patient has relapsed following 

previous treatment with a B cell depleting antibody. 

86. The method of Claim 85 wherein said antibody is Rituxan®. 



-68- 



INTERNATIONAL SEARCH REPORT 



Interr si Application No 

PCT/US 01/10382 



A. CLASSIFICATION OF SUBJECT MATTER , , , , , , 

IPC 7 A61K39/395 A61P35/00 //C07K16/24,C07K16/28,(A61K39/395, 
31:00) 



According lo Inlernalional Patent Classification (IPC) or lo both national classification and IPC 



B. FIELDS SEARCHED ■ 

Minimum documentation searched (classification system followed by classification symbols) 

IPC 7 C07K A61K 



Documentation searched other than minimum documentation to the extent that such documents are included in the fields searched 



Electronic data base consulted during the international search (name of data base and, where practical, search terms used) 

EPO-Internal , WPI Data, PAJ, BIOSIS 



C. DOCUMENTS CONSIDERED TO BE RELEVANT 



Category ° Citation of document, with indication, where appropriate, of the relevant passages 



Relevant to claim No. 



DE 295 12 719 U (MARX UWE DR MED ; SABAT 
ROBERT (DE); GLASER RALF DR RER NAT (DE); 
S) 29 August 1996 (1996-08-29) 
page 2, paragraph 1 
page 3, paragraph 5 



72,73 



1-14, 

18-47, 

50, 

52-56, 
58-71, 
74-86 



-/» 



Further documents are listed in the continuation of box C. 



Patent family members are listed In annex. 



° Special categories of cited documents : 

'A' document defining the general state of the art which is no) 
considered to be of particular relevance 

'E' earlier document but published on or after the international 
filing date 

"L" document which may throw doubts on priority clalm(s) or 
which is cited to establish the publication date of another 
citation or other special reason (as specified) 

"O" document referring to an oral disclosure, use, exhibition or 
other means 

"P* document published prior lo the International filing date but 
later than the priority date claimed 



T later document published after the international filing date 
or priority date and not in conflict with the application but 
cited to understand the principle or theory underlying the 
Invention 

'X' document of particular relevance; the claimed Invention 
cannot be considered novel or cannot be considered to 
involve an inventive step when the document is taken alone 

"Y" document of particular relevance; the claimed Invention 
cannot be considered to involve an Inventive step when the 
document is combined with one or more other such docu- 
ments, such combination being obvious to a person skilled 
In the art. 

'&' document member of the same patent family 



Date of the actual completion of the International search 

28 August 2001 


Date of mailing of the international search report 

10/09/2001 


Name and mailing address of the ISA 

European Patent Office, P.B. 5618 Patentlaan 2 
NL-2280HVRijswtjk 
Tel (+31-70) 340-2040, TX. 31 651 epo nl, 
Fax (+31-70)340-3016 


Authorized officer 

Muller-Ihomalla, K 



Foim PC171SA/210 (second sheat) (July 1892) 



page 1 of 3 



INTERNATIONAL SEARCH REPORT 



Intern 1 Application No 

PCT/US 01/10382 



^Continuation) DOCUMENTS CONSIDERED TO BE RELEVANT 


Category" 


Citation of document, with indlcation,where appropriate, of the relevant passages 


Relevant to claim No. 


X 


WO 94 04180 A (SCHERING CORP) 


72,73 




3 March 1994 (1994-03-03) 






abstract 






page 1, line 9 -page 2, line 9 




Y 


claims 1-11 


1-14, 






18-47, 






50, 






52-56, 






58-71 , 






74-86' 


Y 




CZUCZMAN M S ET AL: "Treatment of 


1-14, 




patients with low-grade B-cell Lymphoma 


18-47, 




with the combination of chimeric anti-CD20 


50, 




monoclonal antibody and CHOP chemotherapy" 


52-56, 




JOURNAL OF CLINICAL ONCOLOGY, 


58-71, 




PHILADELPHIA, PA, US, 


74-86 




vol. 17, no. 1, January 1999 (1999-01), 






pages 268-276, XP000952705 






abstract 






page 268, column 1, line 1 -page 269, 






column 2, paragraph 2 






page 270, column 1, paragraph 3 -column 2, 






paragraph 3 






page 275, column 2; paragraph 2 




Y 


DATABASE BI0SIS 'Online' 

LS (1 1 f \ \J fl W L_> W. A W w X w Will 1 1 1 v • 


29-47, 




BIOSCIENCES INFORMATION SERVICE 


50 




PHILADELPHIA PA US* 1997 


52-56, 




DEMIDEM AICHA ET AL- "Chimeric ant1-CD20 


58-71^ 




(IDEC-C2B8) monoclonal antlbodv sensitizes 


74-86' 




a B cell lymphoma cell line to cell 






killing by cytotoxic drugs." 






Database accession no. PREV199799709997 






XP002176012 






abstract 






& CANCER BIOTHERAPY & 






RADIOPHARMACEUTICALS, 






vol. 12, no. 3, 1997, pages 177-186, 






ISSN: 1084-9785 




Y 


BUSKE C ET AL: "Monoclonal antibody 


29-47, I 




therapy for b cell non-Hodgkin's 


50, 




lymphomas: emerging concepts of a 


52-56, 




tumour-targeted strategy" 


58-71, 




EUROPEAN JOURNAL OF CANCER, PERGAM0N 


74-86 




PRESS, OXFORD, GB, 






vol. 35, no. 4, 1999, pages 549-557, 






XP001010018 






ISSN: 0959-8049 












page 550, column 1, paragraph 4 -page 553, 






column 2, paragraph 1 






page 555, column 1, last paragraph 






_/- 





Foim PCT/1SA/210 (continuation of second shaot) (July 1992) 



page 2 of 3 



INTERNATIONAL SEARCH REPORT lnterr , 1AppllcatlonNo 

PCT/US 01/10382 


C.(Contlnuatlon) DOCUMENTS CONSIDERED TO BE RELEVANT 


Category • 


Citation of document, with Indlcation.where appropriate, of the relevant passages 


Relevant to claim No. 


A 


CORTES J KURZROCK R: "Inter! eukin-10 in 

non-Hodgk1n's lymphoma" 

LEUKEMIA AND LYMPHOMA, HARWOOD ACADEMIC 

PUBLISHERS, CHUR, CH, 

vol. 26, no. 3/4, July 1997 (1997-07), 

pages 251-259, XP001020774 

ISSN: 1042-8194 

the whole document 


1-14, 

18-47, 

50, 

52-56, 
58-86 



Form FCT/1SA/210 (continuation of second shoot) (July 1892) 



page 3 of 3 



INTERNATIONAL SEARCH REPORT 



International Application No. PCTAlS 01 A0382 



FURTHER INFORMATION CONTINUED FROM PCT/ISA/ 210 



Continuation of Box 1.2 

Claims Nos.: 15-17,48,49,51,57 



The kits according to claims 15-17,48,49,51,57 have not been searched, as 
the subject-matter of said claims 1s not defined 1n that the latter lack 
concrete technical features defining the kit 1n questions. An attempt is 
made to define said kits by reference to a result to be achieved. Again, 
this lack of clarity 1n the present case is such as to render a 
meaningful search over the whole of the claimed scope impossible. 

The applicant's attention is drawn to the fact that claims, or parts of 
claims, relating to Inventions in respect of which no international 
search report has been established need not be the subject of an 
international preliminary examination (Rule 66.1(e) PCT). The applicant 
is advised that the EPO policy when acting as an International 
Preliminary Examining Authority is normally not to carry out a 
preliminary examination on matter which has not been searched. This is 
the case Irrespective of whether or not the claims are amended following 
receipt of the search report or during any Chapter II procedure. 



INTERNATIONAL SEARCH REPORT 

Information on patent family members 


Inten u Application No 

PCT/US 01/10382 


Patent document 
cited in search report 


Publication 
date 


Patent family 
member(s) 


Publication 
date 



DE 29512719 U 29-08-1996 NONE 



WO 9404180 A 03-03-1994 



IIC 

Uo 


COCQOC/t 
0000004 


A 


Oft 11 IQOil 

zy— n-iyy4 


AT 
Ml 


1 /\9Q/I7 

lo^y^/ 


T 
1 


lb- uz-iyys 


Al 1 

AU 


O8O0Z8 


n 
D 


A7 no 1 ftft"7 

0/ -08-199/ 


Al 1 

AU 


cai nono 

5010893 


A 


1 c no i ftftii 

ib-03-1994 


r a 
LA 


Ol A OOC1 

Z14Z861 


A 


no no inftji 
03-03-1994 




ioyozo4 


A 


no no inftii 
03-08-1994 


U 


yb00441 


A 

A 


in 1 n 1 ftftC 

18-10-1995 


Ut 


oyoioyzi 


n 

V 


1Z-03-1998 


ut 


6y3it>9Zi 


T 
1 


f\A nc infto 
04-06-1998 


iw 
UK 


o/i933 


T 
1 


oo nn iaao 
£3-09-1998 


CD 

tr 


ft£"71 noo 

00/1933 


A 

A 


Oft ftft IftftC 

ZO-09-1995 


CD 

tr 


nom o.c i 

Uoiuybi 


A 

A 


oo in i nm 
ZZ-10-1997 


CO 

to 


Ol 1 1 7£rt 

ziii/69 


T 

1 


i£ no i ftfto 
16-03-1998 


CT 

r 1 


youoy/ 


A 

A 


A7 ft A 1AAC 

0/-U4-iyyb 


TD 


30ZO&35 


T 
1 


oi n*7 mfto 
31-07-1998 


HK. 


10043ZO 


A 

A 


Oft 11 1 nno 

ZU-1 1-1998 


lji i 
HU 


/oy/o 


A 

A 


OO 11 1 ftftC 

Z8-1 1-1995 


T 1 

1L 


100/Zb 


A 

A 


n£ i o onnn 

06-12-2000 


ID 
JP 


8500362 


T 


16-01-1996 


KR 


170037 


D 

D 


m no i nnn 

01-02-1999 


MX 


9305054 


A 

A 


oo no 1 nn a 

Z8-02-1994 


NO 


950600 


A 


20-04-1995 


NZ 


255757 


A 


27-07-1997 


NZ 


314940 


A 


29-07-1999 


PL 


307566 


A 


29-05-1995 


RU 


2120802 


C 


27-10-1998 


SG 


52248 


A 


28-09-1998 


SK 


21195 


A 


01-10-1996 


ZA 


9306060 


A 


23-05-1994 


US 


6106823 


A 


22-08-2000 


US 


5601815 


A 


11-02-1997 



Form PCTrtSA/210 (patera lemily annex) (July 18S2)